1
|
Shao X, Volk L. PICK1 links KIBRA and AMPA receptor subunit GluA2 in coiled-coil-driven supramolecular complexes. J Biol Chem 2025; 301:108397. [PMID: 40074086 DOI: 10.1016/j.jbc.2025.108397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The human memory-associated protein KIBRA regulates synaptic plasticity and trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors, and is implicated in multiple neuropsychiatric and cognitive disorders. How KIBRA forms complexes with and regulates AMPA receptors remains unclear. Here, we show that KIBRA does not interact directly with the AMPA receptor subunit GluA2, but that protein interacting with C kinase 1 (PICK1), a key regulator of AMPA receptor trafficking, can serve as a bridge between KIBRA and GluA2. In contrast, KIBRA can form a complex with GluA1 independent of PICK1. We identified structural determinants of KIBRA-PICK1-AMPAR complexes by investigating interactions and cellular expression patterns of different combinations of KIBRA and PICK1 domain mutants. We find that the PICK1 BAR domain, a coiled-coil structure, is sufficient for interaction with KIBRA, whereas mutation of the PICK1 BAR domain disrupts KIBRA-PICK1-GluA2 complex formation. In addition, KIBRA recruits PICK1 into large supramolecular complexes, a process which requires KIBRA coiled-coil domains. These findings reveal molecular mechanisms by which KIBRA can organize key synaptic signaling complexes.
Collapse
Affiliation(s)
- Xin Shao
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Lenora Volk
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA; Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, Texas, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA; Peter O'Donnell Jr Brain Institute Investigator, UT Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
2
|
Dunham TL, Wilkerson JR, Johnson RC, Huganir RL, Volk LJ. WWC2 modulates GABA A-receptor-mediated synaptic transmission, revealing class-specific mechanisms of synapse regulation by WWC family proteins. Cell Rep 2024; 43:114841. [PMID: 39388350 PMCID: PMC11913214 DOI: 10.1016/j.celrep.2024.114841] [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: 08/03/2023] [Revised: 07/22/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
The WW and C2 domain-containing protein (WWC2) is implicated in several neurological disorders. Here, we demonstrate that WWC2 interacts with inhibitory, but not excitatory, postsynaptic scaffolds, consistent with prior proteomic identification of WWC2 as a putative component of the inhibitory postsynaptic density. Using mice lacking WWC2 expression in excitatory forebrain neurons, we show that WWC2 suppresses γ-aminobutyric acid type-A receptor (GABAAR) incorporation into the plasma membrane and regulates HAP1 and GRIP1, which form a complex promoting GABAAR recycling to the membrane. Inhibitory synaptic transmission is increased in CA1 pyramidal cells lacking WWC2. Furthermore, unlike the WWC2 homolog KIBRA (kidney/brain protein; WWC1), a key regulator of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking at excitatory synapses, the deletion of WWC2 does not affect synaptic AMPAR expression. In contrast, loss of KIBRA does not affect GABAAR membrane expression. These data reveal synapse class-selective functions for WWC proteins as regulators of ionotropic neurotransmitter receptors and provide insight into mechanisms regulating GABAAR membrane expression.
Collapse
Affiliation(s)
- Thomas L Dunham
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Julia R Wilkerson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lenora J Volk
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Psychiatry UT Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
3
|
Tona R, Inagaki S, Ishibashi Y, Faridi R, Yousaf R, Roux I, Wilson E, Fenollar-Ferrer C, Chien WW, Belyantseva IA, Friedman TB. Interaction between the TBC1D24 TLDc domain and the KIBRA C2 domain is disrupted by two epilepsy-associated TBC1D24 missense variants. J Biol Chem 2024; 300:107725. [PMID: 39214300 PMCID: PMC11465063 DOI: 10.1016/j.jbc.2024.107725] [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: 03/27/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Mutations of human TBC1D24 are associated with deafness, epilepsy, or DOORS syndrome (deafness, onychodystrophy, osteodystrophy, cognitive disability, and seizures). The causal relationships between TBC1D24 variants and the different clinical phenotypes are not understood. Our hypothesis is that phenotypic heterogeneity of missense mutations of TBC1D24 results, in part, from perturbed binding of different protein partners. To discover novel protein partners of TBC1D24, we conducted yeast two-hybrid (Y2H) screen using mouse full-length TBC1D24 as bait. Kidney and brain protein (KIBRA), a scaffold protein encoded by Wwc1, was identified as a partner of TBC1D24. KIBRA functions in the Hippo signaling pathway and is important for human cognition and memory. The TBC1D24 TLDc domain binds to KIBRA full-length and to its C2 domain, confirmed by Y2H assays. No interaction was detected with Y2H assays between the KIBRA C2 domain and TLDc domains of NCOA7, MEAK7, and OXR1. Moreover, the C2 domains of other WWC family proteins do not interact with the TLDc domain of TBC1D24, demonstrating specificity. The mRNAs encoding TBC1D24 and KIBRA proteins in mouse are coexpressed at least in a subset of hippocampal cells indicating availability to interact in vivo. As two epilepsy-associated recessive variants (Gly511Arg and Ala515Val) in the TLDc domain of human TBC1D24 disrupt the interaction with the human KIBRA C2 domain, this study reveals a pathogenic mechanism of TBC1D24-associated epilepsy, linking the TBC1D24 and KIBRA pathways. The interaction of TBC1D24-KIBRA is physiologically meaningful and necessary to reduce the risk of epilepsy.
Collapse
Affiliation(s)
- Risa Tona
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Sayaka Inagaki
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA.
| | - Yasuko Ishibashi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA; Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Rabia Faridi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Rizwan Yousaf
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Isabelle Roux
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA; Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Elizabeth Wilson
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA; Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Wade W Chien
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA; Department of Otolaryngology-Head & Neck Surgery, Johns Hopkins School of Medicine, Maryland, USA
| | - Inna A Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Maryland, USA.
| |
Collapse
|
4
|
Tsokas P, Hsieh C, Flores-Obando RE, Bernabo M, Tcherepanov A, Hernández AI, Thomas C, Bergold PJ, Cottrell JE, Kremerskothen J, Shouval HZ, Nader K, Fenton AA, Sacktor TC. KIBRA anchoring the action of PKMζ maintains the persistence of memory. SCIENCE ADVANCES 2024; 10:eadl0030. [PMID: 38924398 PMCID: PMC11204205 DOI: 10.1126/sciadv.adl0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
How can short-lived molecules selectively maintain the potentiation of activated synapses to sustain long-term memory? Here, we find kidney and brain expressed adaptor protein (KIBRA), a postsynaptic scaffolding protein genetically linked to human memory performance, complexes with protein kinase Mzeta (PKMζ), anchoring the kinase's potentiating action to maintain late-phase long-term potentiation (late-LTP) at activated synapses. Two structurally distinct antagonists of KIBRA-PKMζ dimerization disrupt established late-LTP and long-term spatial memory, yet neither measurably affects basal synaptic transmission. Neither antagonist affects PKMζ-independent LTP or memory that are maintained by compensating PKCs in ζ-knockout mice; thus, both agents require PKMζ for their effect. KIBRA-PKMζ complexes maintain 1-month-old memory despite PKMζ turnover. Therefore, it is not PKMζ alone, nor KIBRA alone, but the continual interaction between the two that maintains late-LTP and long-term memory.
Collapse
Affiliation(s)
- Panayiotis Tsokas
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Changchi Hsieh
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Rafael E. Flores-Obando
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Matteo Bernabo
- Department of Psychology, McGill University, Montreal, Quebec H3A 1G1, Canada
| | - Andrew Tcherepanov
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - A. Iván Hernández
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Christian Thomas
- Internal Medicine D (MedD), Department of Molecular Nephrology, University Hospital of Münster, 48149 Münster, Germany
| | - Peter J. Bergold
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - James E. Cottrell
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Joachim Kremerskothen
- Internal Medicine D (MedD), Department of Molecular Nephrology, University Hospital of Münster, 48149 Münster, Germany
| | - Harel Z. Shouval
- Department of Neurobiology and Anatomy, University of Texas Medical at Houston, Houston, TX 77030, USA
| | - Karim Nader
- Department of Psychology, McGill University, Montreal, Quebec H3A 1G1, Canada
| | - André A. Fenton
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
- Neuroscience Institute at NYU Langone Medical Center, New York, NY 10016, USA
| | - Todd C. Sacktor
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Department of Neurology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| |
Collapse
|
5
|
Shao X, Volk L. PICK1 links KIBRA and AMPA receptors in coiled-coil-driven supramolecular complexes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584494. [PMID: 38558978 PMCID: PMC10980033 DOI: 10.1101/2024.03.12.584494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The human memory-associated protein KIBRA regulates synaptic plasticity and trafficking of AMPA-type glutamate receptors, and is implicated in multiple neuropsychiatric and cognitive disorders. How KIBRA forms complexes with and regulates AMPA receptors remains unclear. Here, we show that KIBRA does not interact directly with the AMPA receptor subunit GluA2, but that PICK1, a key regulator of AMPA receptor trafficking, can serve as a bridge between KIBRA and GluA2. We identified structural determinants of KIBRA-PICK1-AMPAR complexes by investigating interactions and cellular expression patterns of different combinations of KIBRA and PICK1 domain mutants. We find that the PICK1 BAR domain, a coiled-coil structure, is sufficient for interaction with KIBRA, whereas mutation of the BAR domain disrupts KIBRA-PICK1-GluA2 complex formation. In addition, KIBRA recruits PICK1 into large supramolecular complexes, a process which requires KIBRA coiled-coil domains. These findings reveal molecular mechanisms by which KIBRA can organize key synaptic signaling complexes.
Collapse
|
6
|
Dunham TL, Wilkerson JR, Johnson RC, Huganir RL, Volk LJ. Modulation of GABA A receptor trafficking by WWC2 reveals class-specific mechanisms of synapse regulation by WWC family proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584487. [PMID: 38559047 PMCID: PMC10979870 DOI: 10.1101/2024.03.11.584487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
WWC2 (WW and C2 domain-containing protein) is implicated in several neurological disorders, however its function in the brain has yet to be determined. Here, we demonstrate that WWC2 interacts with inhibitory but not excitatory postsynaptic scaffolds, consistent with prior proteomic identification of WWC2 as a putative component of the inhibitory postsynaptic density. Using mice lacking WWC2 expression in excitatory forebrain neurons, we show that WWC2 suppresses GABA A R incorporation into the plasma membrane and regulates HAP1 and GRIP1, which form a complex promoting GABA A R recycling to the membrane. Inhibitory synaptic transmission is dysregulated in CA1 pyramidal cells lacking WWC2. Furthermore, unlike the WWC2 homolog KIBRA (WWC1), a key regulator of AMPA receptor trafficking at excitatory synapses, deletion of WWC2 does not affect synaptic AMPAR expression. In contrast, loss of KIBRA does not affect GABA A R membrane expression. These data reveal unique, synapse class-selective functions for WWC proteins as regulators of ionotropic neurotransmitter receptors and provide insight into mechanisms regulating GABA A R membrane expression.
Collapse
|
7
|
Huang H, Gu J, Kuang X, Yu Y, Rao B, Fang S, Lu J, Qiu F. An integrative pan-cancer analysis of WWC family genes and functional validation in lung cancer. Cell Signal 2024; 115:111034. [PMID: 38190957 DOI: 10.1016/j.cellsig.2024.111034] [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: 09/16/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/10/2024]
Abstract
The WW and C2 domain containing (WWC) protein family functions as scaffolds regulating cell proliferation and organ growth control through the Hippo signaling pathway. However, their pan-cancer dysregulation and mechanistic roles in signaling transduction have remained unclear. We performed integrated pan-cancer analyses of WWC family gene expression using data from The Cancer Genome Atlas (TCGA) across 33 different cancer types. Prognostic relevance was evaluated by survival analyses. WWC genetic alterations, DNA methylation, pathway activities, drug response, and tumor immunology were analyzed using online databases. Furthermore, we examined the functional roles of WWCs in lung cancer cells. We observed aberrant WWC expression in various cancers, which associated with patient prognosis. WWC hypermethylation occurred in many cancers and exhibited negative correlation with expression, alongside mutations linked to poor outcomes. Pathway analysis implicated WWCs as Hippo pathway scaffolds, while drug sensitivity analysis suggested associations with diverse chemotherapies. Additionally, pan-cancer analyses elucidated vital immunomodulatory roles for WWC through heterogeneous correlations with immune cell infiltrates, checkpoint molecules, tumor mutation burden, microsatellite instability, and chemokine pathways across cancers. Experimentally, WWCs suppressed lung cancer cell proliferation, migration, and invasion while enhancing apoptosis and paclitaxel chemosensitivity. Mechanistically, WWCs bound large tumor suppressor 1 and 2 (LATS1/2) kinases to stimulate phosphorylation cascades, thereby inhibiting nuclear translocation of the Yes-associated protein (YAP) oncoprotein. Taken together, our multi-omics characterization provides comprehensive evidence for WWCs as putative tumor suppressors across cancers via Hippo pathway modulation. WWCs may serve as prognostic markers and therapeutic targets in lung cancer.
Collapse
Affiliation(s)
- Hongmei Huang
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Jiaji Gu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Xinjie Kuang
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Yonghui Yu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Boqi Rao
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Shenying Fang
- The fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, PR China
| | - Jiachun Lu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Fuman Qiu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China.
| |
Collapse
|
8
|
Kauwe G, Pareja-Navarro KA, Yao L, Chen JH, Wong I, Saloner R, Cifuentes H, Nana AL, Shah S, Li Y, Le D, Spina S, Grinberg LT, Seeley WW, Kramer JH, Sacktor TC, Schilling B, Gan L, Casaletto KB, Tracy TE. KIBRA repairs synaptic plasticity and promotes resilience to tauopathy-related memory loss. J Clin Invest 2024; 134:e169064. [PMID: 38299587 PMCID: PMC10836803 DOI: 10.1172/jci169064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/21/2023] [Indexed: 02/02/2024] Open
Abstract
Synaptic plasticity is obstructed by pathogenic tau in the brain, representing a key mechanism that underlies memory loss in Alzheimer's disease (AD) and related tauopathies. Here, we found that reduced levels of the memory-associated protein KIdney/BRAin (KIBRA) in the brain and increased KIBRA protein levels in cerebrospinal fluid are associated with cognitive impairment and pathological tau levels in disease. We next defined a mechanism for plasticity repair in vulnerable neurons using the C-terminus of the KIBRA protein (CT-KIBRA). We showed that CT-KIBRA restored plasticity and memory in transgenic mice expressing pathogenic human tau; however, CT-KIBRA did not alter tau levels or prevent tau-induced synapse loss. Instead, we found that CT-KIBRA stabilized the protein kinase Mζ (PKMζ) to maintain synaptic plasticity and memory despite tau-mediated pathogenesis. Thus, our results distinguished KIBRA both as a biomarker of synapse dysfunction and as the foundation for a synapse repair mechanism to reverse cognitive impairment in tauopathy.
Collapse
Affiliation(s)
- Grant Kauwe
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Lei Yao
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jackson H. Chen
- Buck Institute for Research on Aging, Novato, California, USA
| | - Ivy Wong
- Buck Institute for Research on Aging, Novato, California, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Helen Cifuentes
- Buck Institute for Research on Aging, Novato, California, USA
| | - Alissa L. Nana
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, California, USA
| | - Yaqiao Li
- Gladstone Institutes, San Francisco, Califoria, USA
| | - David Le
- Gladstone Institutes, San Francisco, Califoria, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Todd C. Sacktor
- The Robert F. Furchgott Center of Neural and Behavioral Science, Departments of Physiology and Pharmacology, Anesthesiology, and Neurology, State University of New York Health Sciences University, Brooklyn, New York, USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, California, USA
| |
Collapse
|
9
|
Cao R, Zhu R, Sha Z, Qi S, Zhong Z, Zheng F, Lei Y, Tan Y, Zhu Y, Wang Y, Wang Y, Yu FX. WWC1/2 regulate spinogenesis and cognition in mice by stabilizing AMOT. Cell Death Dis 2023; 14:491. [PMID: 37528078 PMCID: PMC10394084 DOI: 10.1038/s41419-023-06020-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 07/16/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
WWC1 regulates episodic learning and memory, and genetic nucleotide polymorphism of WWC1 is associated with neurodegenerative diseases such as Alzheimer's disease. However, the molecular mechanism through which WWC1 regulates neuronal function has not been fully elucidated. Here, we show that WWC1 and its paralogs (WWC2/3) bind directly to angiomotin (AMOT) family proteins (Motins), and recruit USP9X to deubiquitinate and stabilize Motins. Deletion of WWC genes in different cell types leads to reduced protein levels of Motins. In mice, neuron-specific deletion of Wwc1 and Wwc2 results in reduced expression of Motins and lower density of dendritic spines in the cortex and hippocampus, in association with impaired cognitive functions such as memory and learning. Interestingly, ectopic expression of AMOT partially rescues the neuronal phenotypes associated with Wwc1/2 deletion. Thus, WWC proteins modulate spinogenesis and cognition, at least in part, by regulating the protein stability of Motins.
Collapse
Affiliation(s)
- Runyi Cao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rui Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhao Sha
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Sixian Qi
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fengyun Zheng
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yubin Lei
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanfeng Tan
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuwen Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, National Children's Medical Center, No. 399 Wanyuan Road, Shanghai, 201102, China.
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, the State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Liu D, Xing Z, Huang J, Schwieter JW, Liu H. Genetic bases of language control in bilinguals: Evidence from an EEG study. Hum Brain Mapp 2023; 44:3624-3643. [PMID: 37051723 PMCID: PMC10203802 DOI: 10.1002/hbm.26301] [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: 10/19/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Previous studies have debated whether the ability for bilinguals to mentally control their languages is a consequence of their experiences switching between languages or whether it is a specific, yet highly-adaptive, cognitive ability. The current study investigates how variations in the language-related gene FOXP2 and executive function-related genes COMT, BDNF, and Kibra/WWC1 affect bilingual language control during two phases of speech production, namely the language schema phase (i.e., the selection of one language or another) and lexical response phase (i.e., utterance of the target). Chinese-English bilinguals (N = 119) participated in a picture-naming task involving cued language switches. Statistical analyses showed that both genes significantly influenced language control on neural coding and behavioral performance. Specifically, FOXP2 rs1456031 showed a wide-ranging effect on language control, including RTs, F(2, 113) = 4.00, FDR p = .036, and neural coding across three-time phases (N2a: F(2, 113) = 4.96, FDR p = .014; N2b: F(2, 113) = 4.30, FDR p = .028, LPC: F(2, 113) = 2.82, FDR p = .060), while the COMT rs4818 (ts >2.69, FDR ps < .05), BDNF rs6265 (Fs >5.31, FDR ps < .05), and Kibra/WWC1 rs17070145 (ts > -3.29, FDR ps < .05) polymorphisms influenced two-time phases (N2a and N2b). Time-resolved correlation analyses revealed that the relationship between neural coding and cognitive performance is modulated by genetic variations in all four genes. In all, these findings suggest that bilingual language control is shaped by an individual's experience switching between languages and their inherent genome.
Collapse
Affiliation(s)
- Dongxue Liu
- Research Center of Brain and Cognitive NeuroscienceLiaoning Normal UniversityDalianChina
- Key Laboratory of Brain and Cognitive NeuroscienceLiaoning ProvinceDalianChina
- Beijing Key Laboratory of Applied Experimental Psychology, Faculty of PsychologyBeijing Normal UniversityBeijingChina
| | - Zehui Xing
- Research Center of Brain and Cognitive NeuroscienceLiaoning Normal UniversityDalianChina
- Key Laboratory of Brain and Cognitive NeuroscienceLiaoning ProvinceDalianChina
| | - Junjun Huang
- Research Center of Brain and Cognitive NeuroscienceLiaoning Normal UniversityDalianChina
- Key Laboratory of Brain and Cognitive NeuroscienceLiaoning ProvinceDalianChina
| | - John W. Schwieter
- Language Acquisition, Multilingualism, and Cognition Laboratory / Bilingualism Matters @ LaurierWilfrid Laurier UniversityWaterlooCanada
- Department of Linguistics and LanguagesMcMaster UniversityHamiltonCanada
| | - Huanhuan Liu
- Research Center of Brain and Cognitive NeuroscienceLiaoning Normal UniversityDalianChina
- Key Laboratory of Brain and Cognitive NeuroscienceLiaoning ProvinceDalianChina
| |
Collapse
|
11
|
Kauwe G, Pareja-Navarro KA, Yao L, Chen JH, Wong I, Saloner R, Cifuentes H, Nana AL, Shah S, Li Y, Le D, Spina S, Grinberg LT, Seeley WW, Kramer JH, Sacktor TC, Schilling B, Gan L, Casaletto KB, Tracy TE. KIBRA repairs synaptic plasticity and promotes resilience to tauopathy-related memory loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.543777. [PMID: 37398236 PMCID: PMC10312627 DOI: 10.1101/2023.06.12.543777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Synaptic plasticity is obstructed by pathogenic tau in the brain, representing a key mechanism that underlies memory loss in Alzheimer's disease (AD) and related tauopathies. Here, we define a mechanism for plasticity repair in vulnerable neurons using the C-terminus of the KIdney/BRAin (KIBRA) protein (CT-KIBRA). We show that CT-KIBRA restores plasticity and memory in transgenic mice expressing pathogenic human tau; however, CT-KIBRA did not alter tau levels or prevent tau-induced synapse loss. Instead, we find that CT-KIBRA binds to and stabilizes protein kinase Mζ (PKMζ) to maintain synaptic plasticity and memory despite tau-mediated pathogenesis. In humans we find that reduced KIBRA in brain and increased KIBRA in cerebrospinal fluid are associated with cognitive impairment and pathological tau levels in disease. Thus, our results distinguish KIBRA both as a novel biomarker of synapse dysfunction in AD and as the foundation for a synapse repair mechanism to reverse cognitive impairment in tauopathy.
Collapse
Affiliation(s)
- Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Lei Yao
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Ivy Wong
- Buck Institute for Research on Aging, Novato, CA USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | | | - Alissa L. Nana
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, CA USA
| | - Yaqiao Li
- Gladstone Institutes, San Francisco, CA USA
| | - David Le
- Gladstone Institutes, San Francisco, CA USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
- Weill Institute for Neurosciences, Department of Pathology, University of California, San Francisco USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
- Weill Institute for Neurosciences, Department of Pathology, University of California, San Francisco USA
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Todd C. Sacktor
- The Robert F. Furchgott Center of Neural and Behavioral Science, Departments of Physiology and Pharmacology, Anesthesiology, and Neurology, State University of New York Health Sciences University, Brooklyn, NY USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, CA USA
| |
Collapse
|
12
|
Höffken V, Di Persio S, Laurentino S, Wyrwoll MJ, Terwort N, Hermann A, Röpke A, Oud MS, Wistuba J, Kliesch S, Pavenstädt HJ, Tüttelmann F, Neuhaus N, Kremerskothen J. WWC2 expression in the testis: Implications for spermatogenesis and male fertility. FASEB J 2023; 37:e22912. [PMID: 37086090 DOI: 10.1096/fj.202200960r] [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: 06/23/2022] [Revised: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 04/23/2023]
Abstract
The family of WWC proteins is known to regulate cell proliferation and organ growth control via the Hippo signaling pathway. As WWC proteins share a similar domain structure and a common set of interacting proteins, they are supposed to fulfill compensatory functions in cells and tissues. While all three WWC family members WWC1, WWC2, and WWC3 are found co-expressed in most human organs including lung, brain, kidney, and liver, in the testis only WWC2 displays a relatively high expression. In this study, we investigated the testicular WWC2 expression in spermatogenesis and male fertility. We show that the Wwc2 mRNA expression level in mouse testes is increased during development in parallel with germ cell proliferation and differentiation. The cellular expression of each individual WWC family member was evaluated in published single-cell mRNA datasets of murine and human testes demonstrating a high WWC2 expression predominantly in early spermatocytes. In line with this, immunohistochemistry revealed cytosolic WWC2 protein expression in primary spermatocytes from human testes displaying full spermatogenesis. In accordance with these findings, markedly lower WWC2 expression levels were detected in testicular tissues from mice and men lacking germ cells. Finally, analysis of whole-exome sequencing data of male patients affected by infertility and unexplained severe spermatogenic failure revealed several heterozygous, rare WWC2 gene variants with a proposed damaging function and putative impact on WWC2 protein structure. Taken together, our findings provide novel insights into the testicular expression of WWC2 and show its cell-specific expression in spermatocytes. As rare WWC2 variants were identified in the background of disturbed spermatogenesis, WWC2 may be a novel candidate gene for male infertility.
Collapse
Affiliation(s)
- Verena Höffken
- Institute of Molecular Nephrology, Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Sandra Laurentino
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Margot J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Nicole Terwort
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Anke Hermann
- Institute of Molecular Nephrology, Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Albrecht Röpke
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - Manon S Oud
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joachim Wistuba
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Hermann J Pavenstädt
- Institute of Molecular Nephrology, Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Joachim Kremerskothen
- Institute of Molecular Nephrology, Internal Medicine D, University Hospital Münster, Münster, Germany
| |
Collapse
|
13
|
Brauer BL, Wiredu K, Gerber SA, Kettenbach AN. Evaluation of Quantification and Normalization Strategies for Phosphoprotein Phosphatase Affinity Proteomics: Application to Breast Cancer Signaling. J Proteome Res 2023; 22:47-61. [PMID: 36448918 PMCID: PMC10625046 DOI: 10.1021/acs.jproteome.2c00465] [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] [Indexed: 12/02/2022]
Abstract
Accurate quantification of proteomics data is essential for revealing and understanding biological signaling processes. We have recently developed a chemical proteomic strategy termed phosphatase inhibitor beads and mass spectrometry (PIB-MS) to investigate endogenous phosphoprotein phosphatase (PPP) dephosphorylation signaling. Here, we compare the robustness and reproducibility of status quo quantification methods for optimal performance and ease of implementation. We then apply PIB-MS to an array of breast cancer cell lines to determine differences in PPP signaling between subtypes. Breast cancer, a leading cause of cancer death in women, consists of three main subtypes: estrogen receptor-positive (ER+), human epidermal growth factor receptor two positive (HER2+), and triple-negative (TNBC). Although there are effective treatment strategies for ER+ and HER2+ subtypes, tumors become resistant and progress. Furthermore, TNBC has few targeted therapies. Therefore, there is a need to identify new approaches for treating breast cancers. Using PIB-MS, we distinguished TNBC from non-TNBC based on subtype-specific PPP holoenzyme composition. In addition, we identified an increase in PPP interactions with Hippo pathway proteins in TNBC. These interactions suggest that phosphatases in TNBC play an inhibitory role on the Hippo pathway and correlate with increased expression of YAP/TAZ target genes both in TNBC cell lines and in TNBC patients.
Collapse
Affiliation(s)
- Brooke L. Brauer
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Kwame Wiredu
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Scott A. Gerber
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Arminja N. Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH
| |
Collapse
|
14
|
Mendoza ML, Quigley LD, Dunham T, Volk LJ. KIBRA regulates activity-induced AMPA receptor expression and synaptic plasticity in an age-dependent manner. iScience 2022; 25:105623. [PMID: 36465112 PMCID: PMC9713372 DOI: 10.1016/j.isci.2022.105623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
A growing body of human literature implicates KIBRA in memory and neurodevelopmental disorders. Memory and the cellular substrates supporting adaptive cognition change across development. Using an inducible KIBRA knockout mouse, we demonstrate that adult-onset deletion of KIBRA in forebrain neurons impairs long-term spatial memory and long-term potentiation (LTP). These LTP deficits correlate with adult-selective decreases in extrasynaptic AMPA receptors under basal conditions, and we identify a role for KIBRA in LTP-induced AMPAR upregulation. In contrast, juvenile-onset deletion of KIBRA in forebrain neurons did not affect LTP and had minimal effects on basal AMPAR expression. LTP did not increase AMPAR protein expression in juvenile WT mice, providing a potential explanation for juvenile resilience to KIBRA deletion. These data suggest that KIBRA serves a unique role in adult hippocampal function through regulation of basal and activity-dependent AMPAR proteostasis that supports synaptic plasticity.
Collapse
Affiliation(s)
- Matthew L. Mendoza
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lilyana D. Quigley
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas Dunham
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lenora J. Volk
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O’ Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Wang K, Li B, Ge L, Xie Y. Molecular insight into the systematic affinity and selectivity of partner recognition sites between the WW1 and WW2 domains of human KIBRA neuroprotein. J Mol Graph Model 2022; 116:108258. [PMID: 35810735 DOI: 10.1016/j.jmgm.2022.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 05/22/2022] [Accepted: 06/21/2022] [Indexed: 12/14/2022]
Abstract
Human KIBRA, a member of the WWC family proteins, is an upstream regulator of the Salvador/Warts/Hippo (SWH) signaling pathway and predominately expressed in nervous system. The protein has two functionally regulatory domains WW1 and WW2 at N-terminal region, which recognize and bind to the PY-motif segments of their partner proteins to serve as a signaling scaffold role in the SWH pathway. The two domains are highly conserved, but their downstream ligands and biological functions may not be fully consistent. In this study, we attempted to systematically profile the PY-motif affinity to and selectivity between KIBRA WW1 and WW2 domains involved in partner recognition sites. Ontology mining was used to enrich the KIBRA-interacting proteins in literature libraries, from which a variety of PY-motif peptide segments were identified, and their binding behavior to each domain was then analyzed by integrating computational modeling and experimental assay. Most PY-motif peptides were found to interact potently with WW1 and WW2, but they generally only exhibit a moderate or modest selectivity between the two domains. Subsequently, several representative peptides were further examined in detail to elucidate the molecular mechanism underlying their affinity and selectivity. It is revealed that the middle motif region of PY-motif peptides is primarily responsible for the affinity and stability of peptide binding, but only contributes marginally to peptide selectivity. Instead, the N-terminal region and, particularly, C-terminal region of PY-motif peptides play a crucial role in the selectivity. Hydrophobic contacts and hydrogen bonds confer stability and specificity to the domain-peptide interaction, respectively.
Collapse
Affiliation(s)
- Kai Wang
- Department of Anesthesiology, Rizhao People's Hospital, Affiliated to Jining Medical University, Rizhao, 276827, China
| | - Baoqiang Li
- Department of Anesthesiology, Rizhao People's Hospital, Affiliated to Jining Medical University, Rizhao, 276827, China
| | - Lei Ge
- Department of Emergency, Rizhao People's Hospital, Affiliated to Jining Medical University, Rizhao, 276827, China
| | - Yi Xie
- Department of Anesthesiology, Zibo Central Hospital, Affiliated to Binzhou Medical University, Zibo, 255020, China.
| |
Collapse
|
16
|
Song L, Han X, Li Y, Han X, Zhao M, Li C, Wang P, Wang J, Dong Y, Cong L, Han X, Hou T, Liu K, Wang Y, Qiu C, Du Y. Thalamic gray matter volume mediates the association between KIBRA polymorphism and olfactory function among older adults: a population-based study. Cereb Cortex 2022; 33:3664-3673. [PMID: 35972417 PMCID: PMC10068283 DOI: 10.1093/cercor/bhac299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 11/12/2022] Open
Abstract
The kidney and brain expressed protein (KIBRA) rs17070145 polymorphism is associated with both structure and activation of the olfactory cortex. However, no studies have thus far examined whether KIBRA can be linked with olfactory function and whether brain structure plays any role in the association. We addressed these questions in a population-based cross-sectional study among rural-dwelling older adults. This study included 1087 participants derived from the Multidomain Interventions to Delay Dementia and Disability in Rural China, who underwent the brain MRI scans in August 2018 to October 2020; of these, 1016 took the 16-item Sniffin' Sticks identification test and 634 (62.40%) were defined with olfactory impairment (OI). Data were analyzed using the voxel-based morphometry analysis and general linear, logistic, and structural equation models. The KIBRA rs17070145 C-allele (CC or CT vs. TT genotype) was significantly associated with greater gray matter volume (GMV) mainly in the bilateral orbitofrontal cortex and left thalamus (P < 0.05) and with the multi-adjusted odds ratio of 0.73 (95% confidence interval 0.56-0.95) for OI. The left thalamic GMV could mediate 8.08% of the KIBRA-olfaction association (P < 0.05). These data suggest that the KIBRA rs17070145 C-allele is associated with a reduced likelihood of OI among older adults, partly mediated through left thalamic GMV.
Collapse
Affiliation(s)
- Lin Song
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Xiaodong Han
- Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Yuanjing Li
- Department of Neurobiology, Care Sciences and Society, Aging Research Center and Center for Alzheimer Research, Karolinska Institutet-Stockholm University, 17177 Stockholm, Sweden
| | - Xiaolei Han
- Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Mingqing Zhao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China
| | - Chunyan Li
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China
| | - Pin Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Jiafeng Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Yi Dong
- Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Xiaojuan Han
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Keke Liu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China.,Department of Neurobiology, Care Sciences and Society, Aging Research Center and Center for Alzheimer Research, Karolinska Institutet-Stockholm University, 17177 Stockholm, Sweden
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong, PR China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, 250021 Jinan, Shandong, PR China
| |
Collapse
|
17
|
Guo CL. Self-Sustained Regulation or Self-Perpetuating Dysregulation: ROS-dependent HIF-YAP-Notch Signaling as a Double-Edged Sword on Stem Cell Physiology and Tumorigenesis. Front Cell Dev Biol 2022; 10:862791. [PMID: 35774228 PMCID: PMC9237464 DOI: 10.3389/fcell.2022.862791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 12/19/2022] Open
Abstract
Organ development, homeostasis, and repair often rely on bidirectional, self-organized cell-niche interactions, through which cells select cell fate, such as stem cell self-renewal and differentiation. The niche contains multiplexed chemical and mechanical factors. How cells interpret niche structural information such as the 3D topology of organs and integrate with multiplexed mechano-chemical signals is an open and active research field. Among all the niche factors, reactive oxygen species (ROS) have recently gained growing interest. Once considered harmful, ROS are now recognized as an important niche factor in the regulation of tissue mechanics and topology through, for example, the HIF-YAP-Notch signaling pathways. These pathways are not only involved in the regulation of stem cell physiology but also associated with inflammation, neurological disorder, aging, tumorigenesis, and the regulation of the immune checkpoint molecule PD-L1. Positive feedback circuits have been identified in the interplay of ROS and HIF-YAP-Notch signaling, leading to the possibility that under aberrant conditions, self-organized, ROS-dependent physiological regulations can be switched to self-perpetuating dysregulation, making ROS a double-edged sword at the interface of stem cell physiology and tumorigenesis. In this review, we discuss the recent findings on how ROS and tissue mechanics affect YAP-HIF-Notch-PD-L1 signaling, hoping that the knowledge can be used to design strategies for stem cell-based and ROS-targeting therapy and tissue engineering.
Collapse
Affiliation(s)
- Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
18
|
Hippo signaling pathway and respiratory diseases. Cell Death Dis 2022; 8:213. [PMID: 35443749 PMCID: PMC9021242 DOI: 10.1038/s41420-022-01020-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022]
Abstract
The hippo signaling pathway is a highly conserved evolutionary signaling pathway that plays an important role in regulating cell proliferation, organ size, tissue development, and regeneration. Increasing evidences consider that the hippo signaling pathway is involved in the process of respiratory diseases. Hippo signaling pathway is mainly composed of mammalian STE20-like kinase 1/2 (MST1/2), large tumor suppressor 1/2 (LATS1/2), WW domain of the Sav family containing protein 1 (SAV1), MOB kinase activator 1 (MOB1), Yes-associated protein (YAP) or transcriptional coactivator with PDZ-binding motif (TAZ), and members of the TEA domain (TEAD) family. YAP is the cascade effector of the hippo signaling pathway. The activation of YAP promotes pulmonary arterial vascular smooth muscle cells (PAVSMCs) proliferation, which leads to pulmonary vascular remodeling; thereby the pulmonary arterial hypertension (PAH) is aggravated. While the loss of YAP leads to high expression of inflammatory genes and the accumulation of inflammatory cells, the pneumonia is consequently exacerbated. In addition, overexpressed YAP promotes the proliferation of lung fibroblasts and collagen deposition; thereby the idiopathic pulmonary fibrosis (IPF) is promoted. Moreover, YAP knockout reduces collagen deposition and the senescence of adult alveolar epithelial cells (AECs); hence the IPF is slowed. In addition, hippo signaling pathway may be involved in the repair of acute lung injury (ALI) by promoting the proliferation and differentiation of lung epithelial progenitor cells and intervening in the repair of pulmonary capillary endothelium. Moreover, the hippo signaling pathway is involved in asthma. In conclusion, the hippo signaling pathway is involved in respiratory diseases. More researches are needed to focus on the molecular mechanisms by which the hippo signaling pathway participates in respiratory diseases.
Collapse
|
19
|
Han X, Wang C, Song L, Wang X, Tang S, Hou T, Liu C, Liang X, Qiu C, Wang Y, Du Y. KIBRA regulates amyloid β metabolism by controlling extracellular vesicles secretion. EBioMedicine 2022; 78:103980. [PMID: 35367771 PMCID: PMC8983338 DOI: 10.1016/j.ebiom.2022.103980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Background Previous research has revealed that KIBRA controls secretion of extracellular vesicles (EVs) by inhibiting the proteasomal degradation of Rab27a and EVs play an important role in amyloid β (Aβ) metabolism and transmission during Alzheimer's disease (AD) pathogenesis. Here, we further test the hypothesis that KIBRA regulates Aβ metabolism via the endosomal-lysosomal system. Methods We generated KIBRA knockout mice on a 5XFAD background and KIBRA knockdown cells in murine HT22 cells with stably overexpressing APP. Various forms of Aβ and quantification of EVs were analyzed by biochemical methods and nanoparticle tracking analysis, respectively. Multivesicular bodies (MVBs) were visualized by electron microscopy and confocal fluorescent microscopy. In a population-based cohort (n = 1419), KIBRA genotypes and plasma Aβ levels were analyzed using multiple-PCR amplification and Simoa, respectively. Findings Multiple forms of Aβ were dramatically attenuated in KIBRA knockout mouse brain, including monomers, oligomers, and extracellular deposition, but KIBRA knockout had no effect on intraneuronal APP C-terminal fragment β (APP-CTFβ)/Aβ levels. KIBRA depletion also decreased APP-CTFβ/Aβ-associated EVs secretion and subsequently enhanced MVBs number. Furthermore, we found that excessive accumulation of MVBs harboring APP-CTFβ/Aβ promoted the MVBs-lysosome fusion for degradation and inhibition of lysosomal function rescued secretion of APP-CTFβ/Aβ-associated EVs. More importantly, whole exon sequencing of KIBRA in a large population-based cohort identified the association of KIBRA rs28421695 polymorphism with plasma Aβ levels. Interpretation These results demonstrate that KIBRA regulates Aβ metabolism via controlling the secretion of APP-CTFβ/Aβ-associated EVs. Funding National Key R&D Program of China, and National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Xiaolei Han
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China
| | - Chaoqun Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China
| | - Lin Song
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, PR China
| | - Xiaojie Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, PR China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, PR China
| | - Cuicui Liu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, PR China
| | - Xiaoyan Liang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China; Department of Neurobiology, Care Sciences and Society, Aging Research Center and Center for Alzheimer Research, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, PR China.
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Shandong University, No. 324 Jingwuweiqi Road, Jinan, Shandong 250021, PR China; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, PR China.
| |
Collapse
|
20
|
Brücher VC, Egbring C, Plagemann T, Nedvetsky PI, Höffken V, Pavenstädt H, Eter N, Kremerskothen J, Heiduschka P. Lack of WWC2 Protein Leads to Aberrant Angiogenesis in Postnatal Mice. Int J Mol Sci 2021; 22:5321. [PMID: 34070186 PMCID: PMC8158494 DOI: 10.3390/ijms22105321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/03/2023] Open
Abstract
The WWC protein family is an upstream regulator of the Hippo signalling pathway that is involved in many cellular processes. We examined the effect of an endothelium-specific WWC1 and/or WWC2 knock-out on ocular angiogenesis. Knock-outs were induced in C57BL/6 mice at the age of one day (P1) and evaluated at P6 (postnatal mice) or induced at the age of five weeks and evaluated at three months of age (adult mice). We analysed morphology of retinal vasculature in retinal flat mounts. In addition, in vivo imaging and functional testing by electroretinography were performed in adult mice. Adult WWC1/2 double knock-out mice differed neither functionally nor morphologically from the control group. In contrast, the retinas of the postnatal WWC knock-out mice showed a hyperproliferative phenotype with significantly enlarged areas of sprouting angiogenesis and a higher number of tip cells. The branching and end points in the peripheral plexus were significantly increased compared to the control group. The deletion of the WWC2 gene was decisive for these effects; while knocking out WWC1 showed no significant differences. The results hint strongly that WWC2 is an essential regulator of ocular angiogenesis in mice. As an activator of the Hippo signalling pathway, it prevents excessive proliferation during physiological angiogenesis. In adult animals, WWC proteins do not seem to be important for the maintenance of the mature vascular plexus.
Collapse
Affiliation(s)
- Viktoria Constanze Brücher
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Charlotte Egbring
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Tanja Plagemann
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Pavel I. Nedvetsky
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Verena Höffken
- Medical Cell Biology, Medical Clinic D, University of Münster Medical School, 48149 Münster, Germany;
| | - Hermann Pavenstädt
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Nicole Eter
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Joachim Kremerskothen
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Peter Heiduschka
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| |
Collapse
|
21
|
The Hippo pathway component Wwc2 is a key regulator of embryonic development and angiogenesis in mice. Cell Death Dis 2021; 12:117. [PMID: 33483469 PMCID: PMC7822818 DOI: 10.1038/s41419-021-03409-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/27/2020] [Accepted: 01/07/2021] [Indexed: 01/30/2023]
Abstract
The WW-and-C2-domain-containing (WWC) protein family is involved in the regulation of cell differentiation, cell proliferation, and organ growth control. As upstream components of the Hippo signaling pathway, WWC proteins activate the Large tumor suppressor (LATS) kinase that in turn phosphorylates Yes-associated protein (YAP) and its paralog Transcriptional coactivator-with-PDZ-binding motif (TAZ) preventing their nuclear import and transcriptional activity. Inhibition of WWC expression leads to downregulation of the Hippo pathway, increased expression of YAP/TAZ target genes and enhanced organ growth. In mice, a ubiquitous Wwc1 knockout (KO) induces a mild neurological phenotype with no impact on embryogenesis or organ growth. In contrast, we could show here that ubiquitous deletion of Wwc2 in mice leads to early embryonic lethality. Wwc2 KO embryos display growth retardation, a disturbed placenta development, impaired vascularization, and finally embryonic death. A whole-transcriptome analysis of embryos lacking Wwc2 revealed a massive deregulation of gene expression with impact on cell fate determination, cell metabolism, and angiogenesis. Consequently, a perinatal, endothelial-specific Wwc2 KO in mice led to disturbed vessel formation and vascular hypersprouting in the retina. In summary, our data elucidate a novel role for Wwc2 as a key regulator in early embryonic development and sprouting angiogenesis in mice.
Collapse
|
22
|
Höffken V, Hermann A, Pavenstädt H, Kremerskothen J. WWC Proteins: Important Regulators of Hippo Signaling in Cancer. Cancers (Basel) 2021; 13:cancers13020306. [PMID: 33467643 PMCID: PMC7829927 DOI: 10.3390/cancers13020306] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The conserved Hippo pathway regulates cell proliferation and apoptosis via a complex interplay of transcriptional activities, post-translational protein modifications, specific protein–protein interactions and cellular transport processes. Deregulating this highly balanced system can lead to hyperproliferation, organ overgrowth and cancer. Although WWC proteins are known as components of the Hippo signaling pathway, their association with tumorigenesis is often neglected. This review aims to summarize the current knowledge on WWC proteins and their contribution to Hippo signaling in the context of cancer. Abstract The Hippo signaling pathway is known to regulate cell differentiation, proliferation and apoptosis. Whereas activation of the Hippo signaling pathway leads to phosphorylation and cytoplasmic retention of the transcriptional coactivator YAP, decreased Hippo signaling results in nuclear import of YAP and subsequent transcription of pro-proliferative genes. Hence, a dynamic and precise regulation of the Hippo signaling pathway is crucial for organ size control and the prevention of tumor formation. The transcriptional activity of YAP is controlled by a growing number of upstream regulators including the family of WWC proteins. WWC1, WWC2 and WWC3 represent cytosolic scaffolding proteins involved in intracellular transport processes and different signal transduction pathways. Earlier in vitro experiments demonstrated that WWC proteins positively regulate the Hippo pathway via the activation of large tumor suppressor kinases 1/2 (LATS1/2) kinases and the subsequent cytoplasmic accumulation of phosphorylated YAP. Later, reduced WWC expression and subsequent high YAP activity were shown to correlate with the progression of human cancer in different organs. Although the function of WWC proteins as upstream regulators of Hippo signaling was confirmed in various studies, their important role as tumor modulators is often overlooked. This review has been designed to provide an update on the published data linking WWC1, WWC2 and WWC3 to cancer, with a focus on Hippo pathway-dependent mechanisms.
Collapse
|
23
|
Sarmasti Emami S, Zhang D, Yang X. Interaction of the Hippo Pathway and Phosphatases in Tumorigenesis. Cancers (Basel) 2020; 12:E2438. [PMID: 32867200 PMCID: PMC7564220 DOI: 10.3390/cancers12092438] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 01/05/2023] Open
Abstract
The Hippo pathway is an emerging tumor suppressor signaling pathway involved in a wide range of cellular processes. Dysregulation of different components of the Hippo signaling pathway is associated with a number of diseases including cancer. Therefore, identification of the Hippo pathway regulators and the underlying mechanism of its regulation may be useful to uncover new therapeutics for cancer therapy. The Hippo signaling pathway includes a set of kinases that phosphorylate different proteins in order to phosphorylate and inactivate its main downstream effectors, YAP and TAZ. Thus, modulating phosphorylation and dephosphorylation of the Hippo components by kinases and phosphatases play critical roles in the regulation of the signaling pathway. While information regarding kinase regulation of the Hippo pathway is abundant, the role of phosphatases in regulating this pathway is just beginning to be understood. In this review, we summarize the most recent reports on the interaction of phosphatases and the Hippo pathway in tumorigenesis. We have also introduced challenges in clarifying the role of phosphatases in the Hippo pathway and future direction of crosstalk between phosphatases and the Hippo pathway.
Collapse
Affiliation(s)
| | | | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (S.S.E.); (D.Z.)
| |
Collapse
|
24
|
Manichaikul A, Peres LC, Wang XQ, Barnard ME, Chyn D, Sheng X, Du Z, Tyrer J, Dennis J, Schwartz AG, Cote ML, Peters E, Moorman PG, Bondy M, Barnholtz-Sloan JS, Terry P, Alberg AJ, Bandera EV, Funkhouser E, Wu AH, Pearce CL, Pike M, Setiawan VW, Haiman CA, Palmer JR, LeMarchand L, Wilkens LR, Berchuck A, Doherty JA, Modugno F, Ness R, Moysich K, Karlan BY, Whittemore AS, McGuire V, Sieh W, Lawrenson K, Gayther S, Sellers TA, Pharoah P, Schildkraut JM. Identification of novel epithelial ovarian cancer loci in women of African ancestry. Int J Cancer 2020; 146:2987-2998. [PMID: 31469419 PMCID: PMC7523187 DOI: 10.1002/ijc.32653] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/29/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
Women of African ancestry have lower incidence of epithelial ovarian cancer (EOC) yet worse survival compared to women of European ancestry. We conducted a genome-wide association study in African ancestry women with 755 EOC cases, including 537 high-grade serous ovarian carcinomas (HGSOC) and 1,235 controls. We identified four novel loci with suggestive evidence of association with EOC (p < 1 × 10-6 ), including rs4525119 (intronic to AKR1C3), rs7643459 (intronic to LOC101927394), rs4286604 (12 kb 3' of UGT2A2) and rs142091544 (5 kb 5' of WWC1). For HGSOC, we identified six loci with suggestive evidence of association including rs37792 (132 kb 5' of follistatin [FST]), rs57403204 (81 kb 3' of MAGEC1), rs79079890 (LOC105376360 intronic), rs66459581 (5 kb 5' of PRPSAP1), rs116046250 (GABRG3 intronic) and rs192876988 (32 kb 3' of GK2). Among the identified variants, two are near genes known to regulate hormones and diseases of the ovary (AKR1C3 and FST), and two are linked to cancer (AKR1C3 and MAGEC1). In follow-up studies of the 10 identified variants, the GK2 region SNP, rs192876988, showed an inverse association with EOC in European ancestry women (p = 0.002), increased risk of ER positive breast cancer in African ancestry women (p = 0.027) and decreased expression of GK2 in HGSOC tissue from African ancestry women (p = 0.004). A European ancestry-derived polygenic risk score showed positive associations with EOC and HGSOC in women of African ancestry suggesting shared genetic architecture. Our investigation presents evidence of variants for EOC shared among European and African ancestry women and identifies novel EOC risk loci in women of African ancestry.
Collapse
Affiliation(s)
- Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Lauren C. Peres
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Xin-Qun Wang
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Mollie E. Barnard
- Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT
| | - Deanna Chyn
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Xin Sheng
- Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA
| | - Zhaohui Du
- Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA
| | - Jonathan Tyrer
- Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Dennis
- Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Ann G. Schwartz
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI
| | - Michele L. Cote
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI
| | - Edward Peters
- Epidemiology Program, Louisiana State University Health Sciences Center School of Public Health, New Orleans, LA
| | - Patricia G. Moorman
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC
| | - Melissa Bondy
- Cancer Prevention and Population Sciences Program, Baylor College of Medicine, Houston, TX
| | - Jill S. Barnholtz-Sloan
- Department of Population and Quantitative Health Sciences and Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Paul Terry
- Department of Medicine, University of Tennessee Medical Center – Knoxville, Knoxville, TN
| | - Anthony J. Alberg
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Elisa V. Bandera
- Department of Population Science, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Ellen Funkhouser
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Anna H. Wu
- Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA
| | - Celeste Leigh Pearce
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Malcom Pike
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | | | - Andrew Berchuck
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC
| | - Jennifer A. Doherty
- Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT
| | - Francesmary Modugno
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA
- Ovarian Cancer Center of Excellence, Womens Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Roberta Ness
- The University of Texas School of Public Health, Houston, TX
| | - Kirsten Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, Ronald Regan UCLA Medical Center, Los Angeles, CA
| | - Alice S. Whittemore
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA
| | - Valerie McGuire
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA
| | - Weiva Sieh
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, NY, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, New York
| | - Kate Lawrenson
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Simon Gayther
- Center for Bioinformatics and Functional Genomics and the Cedars-Sinai Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Thomas A. Sellers
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Paul Pharoah
- Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
25
|
Li C, Niu J, Zhou B, Deng W, Deng F, Zhou Z, Xu G. Dexmedetomidine attenuates cisplatin-induced cognitive impairment by modulating miR-429-3p expression in rats. 3 Biotech 2020; 10:244. [PMID: 32411568 DOI: 10.1007/s13205-020-02217-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/18/2020] [Indexed: 12/15/2022] Open
Abstract
Chemotherapy-induced cognitive impairment (CICI) is widely recognized as a frequent adverse side effect following the administration of chemotherapeutic agents. This study aimed to explore the neuroprotective functions and mechanisms of microRNAs (miRNAs) mediated by dexmedetomidine (Dex) on cisplatin-induced CICI. The model rats received 5 mg/kg cisplatin injections once per week for 4 weeks. Dex (30 μg/kg) was administered before cisplatin treatment. The protective effects of Dex were evaluated using Morris water maze, Nissl staining, and transmission electron microscopy. Dex-mediated miRNAs were screened via miRNA sequencing. The effects of Dex and key miRNAs on mitochondrial DNA gene mt-ND1 and caspase-9 expression were tested. Dex exhibited a protective effect against decreased learning memory ability, hippocampal neuronal damage, and mitochondrial damage in CICI rats. Thirty-nine differentially expressed (DE) miRNAs were screened, 13 of which responded positively to Dex treatment. Gene Ontology annotation identified that DE miRNAs were mainly involved in transcription, DNA-templated. Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that DE miRNAs were mainly involved in neuronal function and brain development-related pathways, such as axon guidance and calcium signaling pathways. Compared to cisplatin treatment, the expression of miR-429-3p responded more strongly to Dex treatment. In cisplatin-treated cultured hippocampal neurons, Dex treatment and miR-429-3p overexpression significantly increased mitochondrial DNA gene mt-ND1expression and decreased caspase-9 expression. Our study suggests that Dex alleviates CICI by modulating miR-429-3p expression in rats. Thus, Dex may be effective in preventing the side effects of cisplatin.
Collapse
|
26
|
Generation and Profiling of Tumor-Homing Induced Neural Stem Cells from the Skin of Cancer Patients. Mol Ther 2020; 28:1614-1627. [PMID: 32402245 DOI: 10.1016/j.ymthe.2020.04.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
The conversion of human fibroblasts into personalized induced neural stem cells (iNSCs) that actively seek out tumors and deliver cytotoxic agents is a highly promising approach for treating various types of cancer. However, the ability to generate iNSCs from the skin of cancer patients has not been explored. Here, we take an important step toward clinical application by generating iNSCs from skin biopsies of human patients undergoing treatment for the aggressive brain cancer, glioblastoma (GBM). We then utilized a panel of functional and genomic studies to investigate the efficacy and tumor-homing capacity of these patient-derived cells, as well as genomic analysis, to characterize the impact of interpatient variability on this personalized cell therapy. From the skin-tissue biopsies, we established fibroblasts and transdifferentiated the cells into iNSCs. Genomic and functional testing revealed marked variability in growth rates, therapeutic agent production, and gene expression during fibroblast-to-iNSC conversion among patient lines. In vivo testing showed patient-derived iNSCs home to tumors, yet rates and expression of homing-related pathways varied among patients. With the use of surgical-resection mouse models of invasive human cluster of differentiation 133+ (CD133+) GBM cells and serial kinetic imaging, we found that "high-performing" patient-derived iNSC lines reduced the volume of GBM cells 60-fold and extended survival from 28 to 45 days. Treatment with "low-performing" patient lines had minimal effect on tumor growth, but the anti-tumor effect could be rescued by increasing the intracavity dose. Together, these data show, for the first time, that tumor-homing iNSCs can be generated from the skin of cancer patients and efficaciously suppress tumor growth. We also begin to define genetic markers that could be used to identify cells that will contain the most effective attributes for tumor homing and kill in human patients, including high gene expression of the semaphorin-3B (SEMA3B), which is known to be involved in neuronal cell migration. These studies should serve as an important guide toward clinical GBM therapy, where the personalized nature of optimized iNSC therapy has the potential to avoid transplant rejection and maximize treatment durability.
Collapse
|
27
|
Wasinski B, Sohail A, Bonfil RD, Kim S, Saliganan A, Polin L, Bouhamdan M, Kim HRC, Prunotto M, Fridman R. Discoidin Domain Receptors, DDR1b and DDR2, Promote Tumour Growth within Collagen but DDR1b Suppresses Experimental Lung Metastasis in HT1080 Xenografts. Sci Rep 2020; 10:2309. [PMID: 32047176 PMCID: PMC7012844 DOI: 10.1038/s41598-020-59028-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
The Discoidin Domain Receptors (DDRs) constitute a unique set of receptor tyrosine kinases that signal in response to collagen. Using an inducible expression system in human HT1080 fibrosarcoma cells, we investigated the role of DDR1b and DDR2 on primary tumour growth and experimental lung metastases. Neither DDR1b nor DDR2 expression altered tumour growth at the primary site. However, implantation of DDR1b- or DDR2-expressing HT1080 cells with collagen I significantly accelerated tumour growth rate, an effect that could not be observed with collagen I in the absence of DDR induction. Interestingly, DDR1b, but not DDR2, completely hindered the ability of HT1080 cells to form lung colonies after intravenous inoculation, suggesting a differential role for DDR1b in primary tumour growth and lung colonization. Analyses of tumour extracts revealed specific alterations in Hippo pathway core components, as a function of DDR and collagen expression, that were associated with stimulation of tumour growth by DDRs and collagen I. Collectively, these findings identified divergent effects of DDRs on primary tumour growth and experimental lung metastasis in the HT1080 xenograft model and highlight the critical role of fibrillar collagen and DDRs in supporting the growth of tumours thriving within a collagen-rich stroma.
Collapse
Affiliation(s)
- Benjamin Wasinski
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Anjum Sohail
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - R Daniel Bonfil
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Pathology, College of Medical Sciences and Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Seongho Kim
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Allen Saliganan
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Mohamad Bouhamdan
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Hyeong-Reh C Kim
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Marco Prunotto
- Hoffmann-La Roche, Basel, Switzerland.,School of Pharmaceutical Sciences, Geneva, Switzerland
| | - Rafael Fridman
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA. .,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, 48201, USA.
| |
Collapse
|
28
|
Isoform Specificity of PKMs during Long-Term Facilitation in Aplysia Is Mediated through Stabilization by KIBRA. J Neurosci 2019; 39:8632-8644. [PMID: 31537706 DOI: 10.1523/jneurosci.0943-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 01/18/2023] Open
Abstract
Persistent activity of protein kinase M (PKM), the truncated form of protein kinase C (PKC), can maintain long-term changes in synaptic strength in many systems, including the hermaphrodite marine mollusk, Aplysia californica Moreover, different types of long-term facilitation (LTF) in cultured Aplysia sensorimotor synapses rely on the activities of different PKM isoforms in the presynaptic sensory neuron and postsynaptic motor neuron. When the atypical PKM isoform is required, the kidney and brain expressed adaptor protein (KIBRA) is also required. Here, we explore how this isoform specificity is established. We find that PKM overexpression in the motor neuron, but not the sensory neuron, is sufficient to increase synaptic strength and that this activity is not isoform-specific. KIBRA is not the rate-limiting step in facilitation since overexpression of KIBRA is neither sufficient to increase synaptic strength, nor to prolong a form of PKM-dependent intermediate synaptic facilitation. However, the isoform specificity of dominant-negative-PKMs to erase LTF is correlated with isoform-specific competition for stabilization by KIBRA. We identify a new conserved region of KIBRA. Different splice isoforms in this region stabilize different PKMs based on the isoform-specific sequence of an α-helix "handle" in the PKMs. Thus, specific stabilization of distinct PKMs by different isoforms of KIBRA can explain the isoform specificity of PKMs during LTF in Aplysia SIGNIFICANCE STATEMENT Long-lasting changes in synaptic plasticity associated with memory formation are maintained by persistent protein kinases. We have previously shown in the Aplysia sensorimotor model that distinct isoforms of persistently active protein kinase Cs (PKMs) maintain distinct forms of long-lasting synaptic changes, even when both forms are expressed in the same motor neuron. Here, we show that, while the effects of overexpression of PKMs are not isoform-specific, isoform specificity is defined by a "handle" helix in PKMs that confers stabilization by distinct splice forms in a previously undefined domain of the adaptor protein KIBRA. Thus, we define new regions in both KIBRA and PKMs that define the isoform specificity for maintaining synaptic strength in distinct facilitation paradigms.
Collapse
|
29
|
Song L, Tang S, Dong L, Han X, Cong L, Dong J, Han X, Zhang Q, Wang Y, Du Y. The Neuroprotection of KIBRA in Promoting Neuron Survival and Against Amyloid β-Induced Apoptosis. Front Cell Neurosci 2019; 13:137. [PMID: 31031595 PMCID: PMC6473163 DOI: 10.3389/fncel.2019.00137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/20/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Recent research has identified the nucleotide polymorphisms of KIdney and BRAin expressed protein (KIBRA) to be associated with cognitive performance, suggesting its vital role in Alzheimer’s disease (AD); however, the underlying molecular mechanism of KIBRA in AD remains obscure. Methods: The AD animal model (APP/PS1 transgenic mice) and KIBRA knockout (KIBRA KO) mice were used to investigate pathophysiological changes of KIBRA in vivo. Mouse hippocampal cell line (HT22) was used to explore its molecular mechanism through KIBRA CRISPR/Cas9-sgRNA system and KIBRA overexpression lentivirus in vitro. Results: Aged APP/PS1 mice displayed increased neuronal apoptosis in the hippocampus, as did KIBRA KO mice. KIBRA deficiency was closely related to neuronal loss in the brain. In addition, knockdown of KIBRA in neuronal cell lines suppressed its growth and elevated apoptosis-associated protein levels under the stress of Aβ1–42 oligomers. On the contrary, overexpression of KIBRA significantly promoted cell proliferation and reduced its apoptosis. Moreover, through screening several survival-related signaling pathways, we found that KIBRA inhibited apoptosis by activating the Akt pathway other than ERK or PKC pathways, which was further confirmed by Akt-specific inhibitor MK2206. Conclusion: Our data indicate that KIBRA may function as a neuroprotective gene in promoting neuron survival and inhibiting Aβ-induced neuronal apoptosis.
Collapse
Affiliation(s)
- Lin Song
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Lingling Dong
- Department of Neurology, Dongying People's Hospital, Dongying, China
| | - Xiaolei Han
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Jixin Dong
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Xiaojuan Han
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Qinghua Zhang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
30
|
KIBRA controls exosome secretion via inhibiting the proteasomal degradation of Rab27a. Nat Commun 2019; 10:1639. [PMID: 30967557 PMCID: PMC6456494 DOI: 10.1038/s41467-019-09720-x] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 03/22/2019] [Indexed: 02/07/2023] Open
Abstract
Exosomes are nanosized membrane vesicles released from cells after fusion of multivesicular bodies (MVBs) with the plasma membrane (PM) and play important roles in intercellular communication and numerous biological processes. However, the molecular mechanisms regulating exosome secretion remain poorly understood. Here we identify KIBRA as an adaptor-like protein that stabilizes Rab27a, which in turn controls exosome secretion both in vitro and in vivo. Knockdown or overexpression of KIBRA in neuronal and podocyte cell lines leads to a decrease or increase of exosome secretion, respectively, and KIBRA depletion increases MVB size and number. Comparing protein profiles between KIBRA knockout and wild-type mouse brain showed significantly decreased Rab27a, a small GTPase that regulates MVB-PM docking. Rab27a is stabilized by interacting with KIBRA, which prevents ubiquitination and degradation via the ubiquitin-proteasome pathway. In conclusion, we show that KIBRA controls exosome secretion via inhibiting the proteasomal degradation of Rab27a. Exosomes are intercellular signaling vesicles created by fusion of multivesicular bodies (MVBs) and the plasma membrane (PM), but secretory regulation is ill-defined. Song et al. show that KIBRA controls exosome secretion by protecting Rab27a from proteasomal degradation, promoting MVB-PM docking.
Collapse
|
31
|
An Y, Zhang Q, Li X, Wang Z, Li Y, Tang X. Upregulated microRNA miR-21 promotes the progression of lung adenocarcinoma through inhibition of KIBRA and the Hippo signaling pathway. Biomed Pharmacother 2018; 108:1845-1855. [DOI: 10.1016/j.biopha.2018.09.125] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/10/2018] [Accepted: 09/23/2018] [Indexed: 12/21/2022] Open
|
32
|
Posner MG, Upadhyay A, Ishima R, Kalli AC, Harris G, Kremerskothen J, Sansom MSP, Crennell SJ, Bagby S. Distinctive phosphoinositide- and Ca 2+-binding properties of normal and cognitive performance-linked variant forms of KIBRA C2 domain. J Biol Chem 2018; 293:9335-9344. [PMID: 29724824 PMCID: PMC6005455 DOI: 10.1074/jbc.ra118.002279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/03/2018] [Indexed: 01/07/2023] Open
Abstract
Kidney- and brain-expressed protein (KIBRA), a multifunctional scaffold protein with around 20 known binding partners, is involved in memory and cognition, organ size control via the Hippo pathway, cell polarity, and membrane trafficking. KIBRA includes tandem N-terminal WW domains, a C2 domain, and motifs for binding atypical PKC and PDZ domains. A naturally occurring human KIBRA variant involving residue changes at positions 734 (Met-to-Ile) and 735 (Ser-to-Ala) within the C2 domain affects cognitive performance. We have elucidated 3D structures and calcium- and phosphoinositide-binding properties of human KIBRA C2 domain. Both WT and variant C2 adopt a canonical type I topology C2 domain fold. Neither Ca2+ nor any other metal ion was bound to WT or variant KIBRA C2 in crystal structures, and Ca2+ titration produced no significant reproducible changes in NMR spectra. NMR and X-ray diffraction data indicate that KIBRA C2 binds phosphoinositides via an atypical site involving β-strands 5, 2, 1, and 8. Molecular dynamics simulations indicate that KIBRA C2 interacts with membranes via primary and secondary sites on the same domain face as the experimentally identified phosphoinositide-binding site. Our results indicate that KIBRA C2 domain association with membranes is calcium-independent and involves distinctive C2 domain-membrane relative orientations.
Collapse
Affiliation(s)
- Mareike G. Posner
- From the Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Abhishek Upadhyay
- From the Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Rieko Ishima
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Antreas C. Kalli
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, United Kingdom, ,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Gemma Harris
- Research Complex at Harwell, Rutherford Appleton Laboratory, Didcot OX11 0FA, United Kingdom
| | - Joachim Kremerskothen
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, D-48149 Münster, Germany, and
| | - Mark S. P. Sansom
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Susan J. Crennell
- From the Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Stefan Bagby
- From the Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom, , To whom correspondence should be addressed. Tel.:
44-1225-386436; Fax:
44-1225-386779; E-mail:
| |
Collapse
|
33
|
Mudduwa L, Peiris H, Gunasekara S, Abeysiriwardhana D, Liyanage N, Rayala SK, Liyanage T. KIBRA; a novel biomarker predicting recurrence free survival of breast cancer patients receiving adjuvant therapy. BMC Cancer 2018; 18:589. [PMID: 29793439 PMCID: PMC5968668 DOI: 10.1186/s12885-018-4491-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 05/08/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND This study was carried out to evaluate the prognostic value of KIBRA in breast cancer. METHODS This retrospective study included breast cancer patients who sought the services of the immunohistochemistry laboratory of our unit from 2006 to 2015. Tissue microarrays were constructed and immunohistochemical staining was done to assess the KIBRA expression. The Kaplan-Meier model for univariate and Cox-regression model with backward stepwise factor retention method for multivariate analyses were used. Chi square test was used to find out the associations with the established prognostic features. RESULTS A total of 1124 patients were included in the study and KIBRA staining of 909 breast cancers were available for analysis. Cytoplasmic KIBRA expression was seen in 39.5% and nuclear expression in 44.8%. Overall KIBRA-low breast cancers accounted for 41.5%. KIBRA nuclear expression was significantly associated with positive ER and PR expression. Luminal breast cancer patients who had endocrine therapy and KIBRA-low expression had a RFS disadvantage over those who were positive for KIBRA (p = 0.02). Similarly, patients who received chemotherapy and had overall KIBRA-low expression also demonstrated a RFS disadvantage compared to those who had overall positive KIBRA expression (p = 0.018). This effect of KIBRA was independent of the other factors considered for the model. CONCLUSION Overall low-KIBRA expression has an independent effect on the RFS and predicts the RFS outcome of luminal breast cancer patients who received endocrine therapy and breast cancer patients who received chemotherapy.
Collapse
Affiliation(s)
- Lakmini Mudduwa
- Department of Pathology, Faculty of Medicine, University of Ruhuna, Galle, 80000, Sri Lanka.
| | - Harshini Peiris
- Medical Laboratory Science Degree Programme, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - Shania Gunasekara
- Department of Pathology, Faculty of Medicine, University of Ruhuna, Galle, 80000, Sri Lanka
| | | | - Nimsha Liyanage
- Department of Pathology, Faculty of Medicine, University of Ruhuna, Galle, 80000, Sri Lanka
| | - Suresh K Rayala
- Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, 600 036, India
| | - Thusharie Liyanage
- Department of Pathology, Faculty of Medicine, University of Ruhuna, Galle, 80000, Sri Lanka
| |
Collapse
|
34
|
Feng J, Dong L, Zhang J, Han X, Tang S, Song L, Cong L, Wang X, Wang Y, Du Y. Unique expression pattern of KIBRA in the enteric nervous system of APP/PS1 mice. Neurosci Lett 2018. [DOI: 10.1016/j.neulet.2018.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Hermann A, Wennmann DO, Gromnitza S, Edeling M, Van Marck V, Sudol M, Schaefer L, Duning K, Weide T, Pavenstädt H, Kremerskothen J. WW and C2 domain-containing proteins regulate hepatic cell differentiation and tumorigenesis through the hippo signaling pathway. Hepatology 2018; 67:1546-1559. [PMID: 29116649 DOI: 10.1002/hep.29647] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/12/2017] [Accepted: 11/03/2017] [Indexed: 12/28/2022]
Abstract
UNLABELLED The Hippo pathway regulates cell differentiation, proliferation, and apoptosis. Upon activation, it inhibits the import of the transcriptional coactivator yes-associated protein (YAP) into the nucleus, thus suppressing transcription of pro-proliferative genes. Hence, dynamic and precise control of the Hippo pathway is crucial for organ size control and the prevention of tumor formation. Hippo signaling is controlled by a growing number of upstream regulators, including WW and C2 domain-containing (WWC) proteins, which trigger a serine/threonine kinase pathway. One component of this is the large tumor suppressor (LATS) kinase, which phosphorylates YAP, trapping it in the cytoplasm. WWC proteins have been shown to interact with LATS in vitro and stimulate its kinase activity, thus directly promoting cytoplasmic accumulation of phosphorylated YAP. However, the function of the WWC proteins in the regulation of cell proliferation, organ size control, and tumor prevention in vivo has not yet been determined. Here, we show that loss of hepatic WWC expression in mice leads to tissue overgrowth, inflammation, fibrosis, and formation of liver carcinoma. WWC-deficient mouse livers display reduced LATS activity, increased YAP-mediated gene transcription, and enhanced proliferation of hepatic progenitor cells. In addition, loss of WWC expression in the liver accelerates the turnover of angiomotin proteins, which act as negative regulators of YAP activity. CONCLUSION Our data define an essential in vivo function for WWC proteins as regulators of canonical and noncanonical Hippo signaling in hepatic cell growth and liver tumorigenesis. Thus, expression of WWC proteins may serve as novel prognostic factors in human liver carcinoma. (Hepatology 2018;67:1546-1559).
Collapse
Affiliation(s)
- Anke Hermann
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Dirk Oliver Wennmann
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Sascha Gromnitza
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Maria Edeling
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Veerle Van Marck
- Institute for Pathology, University Hospital Muenster, Münster, Germany
| | - Marius Sudol
- Mechanobiology Institute and Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Liliana Schaefer
- Institute for Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Kerstin Duning
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Thomas Weide
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Hermann Pavenstädt
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| | - Joachim Kremerskothen
- Division of Internal Medicine, Department of Nephrology, Hypertension and Rheumatology, University Hospital Münster, Münster, Germany
| |
Collapse
|
36
|
Chen B, Liu G. WWC3 inhibits intimal proliferation following vascular injury via the Hippo signaling pathway. Mol Med Rep 2018; 17:5175-5183. [PMID: 29393412 PMCID: PMC5865984 DOI: 10.3892/mmr.2018.8484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 11/28/2017] [Indexed: 01/03/2023] Open
Abstract
The Hippo signaling pathway is involved in the formation and development of the cardiovascular system. In the present study, the effects of WWC family member 3 (WWC3) on vascular smooth muscle cells (VSMCs) following injury were investigated, in addition to the associated mechanisms underlying this process. Platelet-derived growth factor BB (PDGF-BB) was used as a cell injury factor, and rats with balloon injuries were used as a model of carotid intimal injury. Furthermore, the expression levels of WWC3 in VSMCs and arteries post-injury were investigated, in addition to the effect of WWC3 on the proliferation and migration of VSMCs. The results demonstrated that following injury, WWC3 expression was suppressed in VSMCs and the rat carotid artery, and the activity of the Hippo signaling pathway was significantly downregulated. In addition, the expression of YY1-associated protein-1 (YAP) and a number of its downstream target genes, including connective tissue growth factor (CTGF), were enhanced, thus enhancing the proliferation and migration of VSMCs. Knockdown of WWC3 suppressed the levels of large tumor suppressor kinase 1 (LATS1) expression and YAP phosphorylation, and the expression of YAP, CTGF and cyclin E was subsequently enhanced, thus promoting cell proliferation and migration. Similar results were obtained following overexpression of WWC3. Treatment with PDGF-BB was revealed to suppress the proliferation and migration of VSMCs transfected with the WWC3 plasmid, compared with VSMCs transfected with an empty vector. The present study demonstrated that WWC3 may interact with LATS1 in order to upregulate the Hippo signaling pathway via co-immunoprecipitation and enhancement of the phosphorylation of LATS1, in addition to the corresponding suppression of the nuclear import of YAP. However, VSMCs transfected with WWC3 plasmid with a deletion of the WW domain fail to exhibit this effect. These results suggested that WWC3 expression is downregulated in VSMCs during neointimal hyperplasia following injury (PDGF-BB stimulation or balloon injury). WWC3 upregulates the activity of the Hippo signaling pathway, and weakens the proliferation and migration of VSMCs. Furthermore, the results of the present study suggested that WWC3 may interact with LATS1 to promote the phosphorylation of YAP and reduce its nuclear translocation, upregulate the activity of the Hippo pathway, and suppress the proliferation and migration of VSMCs following injury.
Collapse
Affiliation(s)
- Beijia Chen
- Department of Cardiology, The First Affiliated Hospital of The China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Guinan Liu
- Department of Cardiology, The First Affiliated Hospital of The China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
37
|
Wigerius M, Quinn D, Diab A, Clattenburg L, Kolar A, Qi J, Krueger SR, Fawcett JP. The polarity protein Angiomotin p130 controls dendritic spine maturation. J Cell Biol 2018; 217:715-730. [PMID: 29317530 PMCID: PMC5800806 DOI: 10.1083/jcb.201705184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/31/2017] [Accepted: 11/30/2017] [Indexed: 01/06/2023] Open
Abstract
Wigerius et al. identify the polarity protein AMOT-130 as vital for dendritic spine morphogenesis. They show that reduced Lats1 kinase activity in the neonatal brain is required for the recruitment of AMOT-130 to postsynaptic compartments to stabilize dendritic spines. The actin cytoskeleton is essential for the structural changes in dendritic spines that lead to the formation of new synapses. Although the molecular mechanisms underlying spine formation are well characterized, the events that drive spine maturation during development are largely unknown. In this study, we demonstrate that Angiomotin (AMOT-130) is necessary for spine stabilization. AMOT-130 is enriched in mature dendritic spines and functions to stabilize the actin cytoskeleton by coupling F-actin to postsynaptic protein scaffolds. These functions of AMOT are transiently restricted during postnatal development by phosphorylation imposed by the kinase Lats1. Our study proposes that AMOT-130 is essential for normal spine morphogenesis and identifies Lats1 as an upstream regulator in this process. Moreover, our findings may link AMOT-130 loss and the related spine defects to neurological disorders.
Collapse
Affiliation(s)
- Michael Wigerius
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | - Dylan Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Antonios Diab
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | | | - Annette Kolar
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Jiansong Qi
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | - Stefan R Krueger
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - James P Fawcett
- Department of Pharmacology, Dalhousie University, Halifax, Canada .,Department of Surgery, Dalhousie University, Halifax, Canada
| |
Collapse
|
38
|
Hu L, Xu Q, Li J, Wang F, Xu X, Sun Z, Ma X, Liu Y, Wang Q, Wang D. No differences in brain microstructure between young KIBRA-C carriers and non-carriers. Oncotarget 2018; 9:1200-1209. [PMID: 29416687 PMCID: PMC5787430 DOI: 10.18632/oncotarget.23348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/04/2017] [Indexed: 11/25/2022] Open
Abstract
KIBRA rs17070145 polymorphism is associated with variations in memory function and the microstructure of related brain areas. Diffusion kurtosis imaging (DKI) as an extension of diffusion tensor imaging that can provide more information about changes in microstructure, based on the idea that water diffusion in biological tissues is heterogeneous due to structural hindrance and restriction. We used DKI to explore the relationship between KIBRA gene polymorphism and brain microstructure in young adults. We recruited 100 healthy young volunteers, including 53 TT carriers and 47 C allele carriers. No differences were detected between the TT homozygotes and C-allele carriers for any diffusion and kurtosis parameter. These results indicate KIBRA rs17070145 polymorphism likely has little or no effect on brain microstructure in young adults.
Collapse
Affiliation(s)
- Li Hu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Qunxing Xu
- Medical Examination Center, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jizhen Li
- Mental Health Center of Shandong Province, Jinan 250012, China
| | - Feifei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xinghua Xu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Zhiyuan Sun
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiangxing Ma
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yong Liu
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Qing Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
39
|
Stepan J, Anderzhanova E, Gassen NC. Hippo Signaling: Emerging Pathway in Stress-Related Psychiatric Disorders? Front Psychiatry 2018; 9:715. [PMID: 30627107 PMCID: PMC6309125 DOI: 10.3389/fpsyt.2018.00715] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/06/2018] [Indexed: 12/20/2022] Open
Abstract
Discovery of the Hippo pathway and its core components has made a significant impact on our progress in the understanding of organ development, tissue homeostasis, and regeneration. Upon diverse extracellular and intracellular stimuli, Hippo signaling regulates stemness, cell proliferation and apoptosis by a well-conserved signaling cascade, and disruption of these systems has been implicated in cancer as well as metabolic and neurodegenerative diseases. The central role of Hippo signaling in cell biology also results in prominent links to stress-regulated pathways. Genetic variations, epigenetically provoked upregulation of Hippo pathway members and dysregulation of cellular processes implicated in learning and memory, are linked to an increased risk of stress-related psychiatric disorders (SRPDs). In this review, we summarize recent findings, supporting the role of Hippo signaling in SRPDs by canonical and non-canonical Hippo pathway interactions.
Collapse
Affiliation(s)
- Jens Stepan
- Department Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elmira Anderzhanova
- Clinic and Polyclinic of Psychiatry and Psychotherapy, Bonn University Clinic, Bonn, Germany
| | - Nils C Gassen
- Clinic and Polyclinic of Psychiatry and Psychotherapy, Bonn University Clinic, Bonn, Germany
| |
Collapse
|
40
|
Erdős G, Szaniszló T, Pajkos M, Hajdu-Soltész B, Kiss B, Pál G, Nyitray L, Dosztányi Z. Novel linear motif filtering protocol reveals the role of the LC8 dynein light chain in the Hippo pathway. PLoS Comput Biol 2017; 13:e1005885. [PMID: 29240760 PMCID: PMC5746249 DOI: 10.1371/journal.pcbi.1005885] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/28/2017] [Accepted: 11/20/2017] [Indexed: 01/12/2023] Open
Abstract
Protein-protein interactions (PPIs) formed between short linear motifs and globular domains play important roles in many regulatory and signaling processes but are highly underrepresented in current protein-protein interaction databases. These types of interactions are usually characterized by a specific binding motif that captures the key amino acids shared among the interaction partners. However, the computational proteome-level identification of interaction partners based on the known motif is hindered by the huge number of randomly occurring matches from which biologically relevant motif hits need to be extracted. In this work, we established a novel bioinformatic filtering protocol to efficiently explore interaction network of a hub protein. We introduced a novel measure that enabled the optimization of the elements and parameter settings of the pipeline which was built from multiple sequence-based prediction methods. In addition, data collected from PPI databases and evolutionary analyses were also incorporated to further increase the biological relevance of the identified motif hits. The approach was applied to the dynein light chain LC8, a ubiquitous eukaryotic hub protein that has been suggested to be involved in motor-related functions as well as promoting the dimerization of various proteins by recognizing linear motifs in its partners. From the list of putative binding motifs collected by our protocol, several novel peptides were experimentally verified to bind LC8. Altogether 71 potential new motif instances were identified. The expanded list of LC8 binding partners revealed the evolutionary plasticity of binding partners despite the highly conserved binding interface. In addition, it also highlighted a novel, conserved function of LC8 in the upstream regulation of the Hippo signaling pathway. Beyond the LC8 system, our work also provides general guidelines that can be applied to explore the interaction network of other linear motif binding proteins or protein domains. Fine-tuning of many cellular processes relies on weak, transient protein-protein interactions. Such interactions often involve compact functional modules, called short linear motifs (SLiMs) that can bind to specific globular domains. SLiM-mediated interactions can carry out diverse molecular functions by targeting proteins to specific cellular locations, regulating the activity and binding preferences of proteins, or aiding the assembly of macromolecular complexes. The key to the function of SLiMs is their small size and highly flexible nature. At the same time, these properties make their experimental identification challenging. Consequently, only a small portion of SLiM-mediated interactions is currently known. This underlies the importance of novel computational methods that can reliably identify candidate sites involved in binding to linear motif binding domains. Here we present a novel bioinformatic approach that efficiently predicts new binding partners for SLiM-binding domains. We applied this method to the dynein light chain LC8, a protein that was already known to bind many partners in a wide range of organisms. With this method, we not only significantly expanded the interaction network of LC8, but also identified a novel function of LC8 in a highly important pathway controlling organ size in animals.
Collapse
Affiliation(s)
- Gábor Erdős
- MTA-ELTE Lendület Bioinformatics Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Szaniszló
- MTA-ELTE Lendület Bioinformatics Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Mátyás Pajkos
- MTA-ELTE Lendület Bioinformatics Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Borbála Hajdu-Soltész
- MTA-ELTE Lendület Bioinformatics Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Bence Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsanna Dosztányi
- MTA-ELTE Lendület Bioinformatics Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
41
|
Abstract
By using a tau construct with two mimicked acetylation sites as identified in AD brains, Tracy et al. (2016) found that acetylated tau promotes synaptic dysfunction through disruption of postsynaptic KIBRA signaling pathways, actin dynamics, and AMPA receptor trafficking.
Collapse
Affiliation(s)
- Jane C Hettinger
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John R Cirrito
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
42
|
Hu J, Ferguson L, Adler K, Farah CA, Hastings MH, Sossin WS, Schacher S. Selective Erasure of Distinct Forms of Long-Term Synaptic Plasticity Underlying Different Forms of Memory in the Same Postsynaptic Neuron. Curr Biol 2017. [PMID: 28648820 DOI: 10.1016/j.cub.2017.05.081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Generalization of fear responses to non-threatening stimuli is a feature of anxiety disorders. It has been challenging to target maladaptive generalized memories without affecting adaptive memories. Synapse-specific long-term plasticity underlying memory involves the targeting of plasticity-related proteins (PRPs) to activated synapses. If distinct tags and PRPs are used for different forms of plasticity, one could selectively remove distinct forms of memory. Using a stimulation paradigm in which associative long-term facilitation (LTF) occurs at one input and non-associative LTF at another input to the same postsynaptic neuron in an Aplysia sensorimotor preparation, we found that each form of LTF is reversed by inhibiting distinct isoforms of protein kinase M (PKM), putative PRPs, in the postsynaptic neuron. A dominant-negative (dn) atypical PKM selectively reversed associative LTF, while a dn classical PKM selectively reversed non-associative LTF. Although both PKMs are formed from calpain-mediated cleavage of protein kinase C (PKC) isoforms, each form of LTF is sensitive to a distinct dn calpain expressed in the postsynaptic neuron. Associative LTF is blocked by dn classical calpain, whereas non-associative LTF is blocked by dn small optic lobe (SOL) calpain. Interfering with a putative synaptic tag, the adaptor protein KIBRA, which protects the atypical PKM from degradation, selectively erases associative LTF. Thus, the activity of distinct PRPs and tags in a postsynaptic neuron contribute to the maintenance of different forms of synaptic plasticity at separate inputs, allowing for selective reversal of synaptic plasticity and providing a cellular basis for developing therapeutic strategies for selectively reversing maladaptive memories.
Collapse
Affiliation(s)
- Jiangyuan Hu
- Department of Neuroscience, Columbia University Medical Center, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, USA.
| | - Larissa Ferguson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Kerry Adler
- Department of Neuroscience, Columbia University Medical Center, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, USA
| | - Carole A Farah
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Margaret H Hastings
- Department of Psychology, McGill University, Montreal Neurological Institute, Montreal, QC H3A 1B1, Canada
| | - Wayne S Sossin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada; Department of Psychology, McGill University, Montreal Neurological Institute, Montreal, QC H3A 1B1, Canada
| | - Samuel Schacher
- Department of Neuroscience, Columbia University Medical Center, New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
43
|
Willsey AJ, Fernandez TV, Yu D, King RA, Dietrich A, Xing J, Sanders SJ, Mandell JD, Huang AY, Richer P, Smith L, Dong S, Samocha KE, Neale BM, Coppola G, Mathews CA, Tischfield JA, Scharf JM, State MW, Heiman GA. De Novo Coding Variants Are Strongly Associated with Tourette Disorder. Neuron 2017; 94:486-499.e9. [PMID: 28472652 PMCID: PMC5769876 DOI: 10.1016/j.neuron.2017.04.024] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/30/2022]
Abstract
Whole-exome sequencing (WES) and de novo variant detection have proven a powerful approach to gene discovery in complex neurodevelopmental disorders. We have completed WES of 325 Tourette disorder trios from the Tourette International Collaborative Genetics cohort and a replication sample of 186 trios from the Tourette Syndrome Association International Consortium on Genetics (511 total). We observe strong and consistent evidence for the contribution of de novo likely gene-disrupting (LGD) variants (rate ratio [RR] 2.32, p = 0.002). Additionally, de novo damaging variants (LGD and probably damaging missense) are overrepresented in probands (RR 1.37, p = 0.003). We identify four likely risk genes with multiple de novo damaging variants in unrelated probands: WWC1 (WW and C2 domain containing 1), CELSR3 (Cadherin EGF LAG seven-pass G-type receptor 3), NIPBL (Nipped-B-like), and FN1 (fibronectin 1). Overall, we estimate that de novo damaging variants in approximately 400 genes contribute risk in 12% of clinical cases. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- A Jeremy Willsey
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Thomas V Fernandez
- Yale Child Study Center and Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Dongmei Yu
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Robert A King
- Yale Child Study Center and Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Andrea Dietrich
- University of Groningen, University Medical Center Groningen, Department of Child and Adolescent Psychiatry, 9713GZ Groningen, the Netherlands
| | - Jinchuan Xing
- Rutgers, the State University of New Jersey, Department of Genetics and the Human Genetics Institute of New Jersey, Piscataway, NJ 08854, USA
| | - Stephan J Sanders
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey D Mandell
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Alden Y Huang
- Department of Neurology, University of California Los Angeles, Los Angeles, California, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Petra Richer
- Yale Child Study Center and Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA; Sewanee: The University of the South, Sewanee, TN 37383, USA
| | - Louw Smith
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Shan Dong
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kaitlin E Samocha
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Benjamin M Neale
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Giovanni Coppola
- Department of Neurology, University of California Los Angeles, Los Angeles, California, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Carol A Mathews
- Department of Psychiatry, University of Florida School of Medicine, Gainesville, FL 32611, USA
| | - Jay A Tischfield
- Rutgers, the State University of New Jersey, Department of Genetics and the Human Genetics Institute of New Jersey, Piscataway, NJ 08854, USA
| | - Jeremiah M Scharf
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Matthew W State
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Gary A Heiman
- Rutgers, the State University of New Jersey, Department of Genetics and the Human Genetics Institute of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
44
|
Saladi SV, Ross K, Karaayvaz M, Tata PR, Mou H, Rajagopal J, Ramaswamy S, Ellisen LW. ACTL6A Is Co-Amplified with p63 in Squamous Cell Carcinoma to Drive YAP Activation, Regenerative Proliferation, and Poor Prognosis. Cancer Cell 2017; 31:35-49. [PMID: 28041841 PMCID: PMC5225026 DOI: 10.1016/j.ccell.2016.12.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 08/18/2016] [Accepted: 11/30/2016] [Indexed: 12/16/2022]
Abstract
Loss-of-function mutations in SWI/SNF chromatin-remodeling subunit genes are observed in many cancers, but an oncogenic role for SWI/SNF is not well established. Here, we reveal that ACTL6A, encoding an SWI/SNF subunit linked to stem cell and progenitor cell function, is frequently co-amplified and highly expressed together with the p53 family member p63 in head and neck squamous cell carcinoma (HNSCC). ACTL6A and p63 physically interact, cooperatively controlling a transcriptional program that promotes proliferation and suppresses differentiation, in part through activation of the Hippo-YAP pathway via regulators including WWC1. Ectopic ACTL6A/p63 expression promotes tumorigenesis, while ACTL6A expression and YAP activation are highly correlated in primary HNSCC and predict poor patient survival. Thus, ACTL6A and p63 collaborate as oncogenic drivers in HNSCC.
Collapse
Affiliation(s)
- Srinivas Vinod Saladi
- Massachusetts General Hospital Cancer Center, GRJ-904, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth Ross
- Massachusetts General Hospital Cancer Center, GRJ-904, 55 Fruit Street, Boston, MA 02114, USA
| | - Mihriban Karaayvaz
- Massachusetts General Hospital Cancer Center, GRJ-904, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Purushothama R Tata
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Hongmei Mou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Internal Medicine, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, GRJ-904, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, GRJ-904, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Paziewska A, Habior A, Rogowska A, Zych W, Goryca K, Karczmarski J, Dabrowska M, Ambrozkiewicz F, Walewska-Zielecka B, Krawczyk M, Cichoz-Lach H, Milkiewicz P, Kowalik A, Mucha K, Raczynska J, Musialik J, Boryczka G, Wasilewicz M, Ciecko-Michalska I, Ferenc M, Janiak M, Kanikowska A, Stankiewicz R, Hartleb M, Mach T, Grzymislawski M, Raszeja-Wyszomirska J, Wunsch E, Bobinski T, Mikula M, Ostrowski J. A novel approach to genome-wide association analysis identifies genetic associations with primary biliary cholangitis and primary sclerosing cholangitis in Polish patients. BMC Med Genomics 2017; 10:2. [PMID: 28056976 PMCID: PMC5217265 DOI: 10.1186/s12920-016-0239-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 12/15/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) are forms of hepatic autoimmunity, and risk for both diseases has a strong genetic component. This study aimed to define the genetic architecture of PBC and PSC within the Polish population. METHODS Subjects were 443 women with PBC, 120 patients with PSC, and 934 healthy controls recruited from Gastroenterology Departments in various Polish hospitals. Allelotyping employed a pooled-DNA sample-based genome-wide association study (GWAS) approach, using Illumina Human Omni2.5-Exome BeadChips and the following novel selection criteria for risk loci: blocks of at least 10 single nucleotide polymorphisms (SNPs) in strong linkage disequilibrium, where the distance between each adjacent SNP pair in the block was less than 30 kb, and each SNP was associated with disease at a significance level of P < 0.005. A selected index SNP from each block was validated using TaqMan SNP genotyping assays. RESULTS Nineteen and twenty-one SNPs were verified as associated with PBC and PSC, respectively, by individual genotyping; 19 (10/9, PBC/PSC) SNPs reached a stringent (corrected) significance threshold and a further 21 (9/12, PBC/PSC) reached a nominal level of significance (P < 0.05 with odds ratio (OR) > 1.2 or < 0.83), providing suggestive evidence of association. The SNPs mapped to seven (1p31.3, 3q13, 6p21, 7q32.1, 11q23.3, 17q12, 19q13.33) and one (6p21) chromosome region previously associated with PBC and PSC, respectively. The SNP, rs35730843, mapping to the POLR2G gene promoter (P = 1.2 × 10-5, OR = 0.39) demonstrated the highest effect size, and was protective for PBC, whereas for PSC respective SNPs were: rs13191240 in the intron of ADGRB3 gene (P = 0.0095, OR = 0.2) and rs3822659 (P = 0.0051, OR = 0.236) along with rs9686714 (P = 0.00077, OR = 0.2), both located in the WWC1 gene. CONCLUSIONS Our cost-effective GWAS approach followed by individual genotyping confirmed several previously identified associations and discovered new susceptibility loci associated with PBC and/or PSC in Polish patients. However, further functional studies are warranted to understand the roles of these newly identified variants in the development of the two disorders.
Collapse
Affiliation(s)
- Agnieszka Paziewska
- Department of Gastroenterology, Hepatology and Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Andrzej Habior
- Department of Gastroenterology, Hepatology and Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Agnieszka Rogowska
- Department of Gastroenterology, Hepatology and Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Włodzimierz Zych
- Department of Gastroenterology, Hepatology and Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Krzysztof Goryca
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Jakub Karczmarski
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Michalina Dabrowska
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Filip Ambrozkiewicz
- Department of Gastroenterology, Hepatology and Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Bozena Walewska-Zielecka
- Department of Public Health, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Marek Krawczyk
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | | | - Piotr Milkiewicz
- Department of General, Liver and Internal Medicine Unit, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | | | - Krzysztof Mucha
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Raczynska
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Musialik
- Department of Gastroenterology and Hepatology, Medical University of Silesia, Katowice, Poland
| | - Grzegorz Boryczka
- Department of Gastroenterology and Hepatology, Medical University of Silesia, Katowice, Poland
| | - Michal Wasilewicz
- Department of General, Liver and Internal Medicine Unit, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Irena Ciecko-Michalska
- Department of Gastroenterology and Infectious Diseases, Collegium Medicum Jagiellonian University, Krakow, Poland
| | - Malgorzata Ferenc
- Department of Gastroenterology, Provincial Hospital, Olsztyn, Poland
| | - Maria Janiak
- Department of Gastroenterology and Hepatology, Medical University of Gdansk, Gdansk, Poland
| | - Alina Kanikowska
- Department of Internal and Metabolic Diseases and Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Rafal Stankiewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Marek Hartleb
- Department of Gastroenterology and Hepatology, Medical University of Silesia, Katowice, Poland
| | - Tomasz Mach
- Department of Gastroenterology and Infectious Diseases, Collegium Medicum Jagiellonian University, Krakow, Poland
| | - Marian Grzymislawski
- Department of Internal and Metabolic Diseases and Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Raszeja-Wyszomirska
- Department of General, Liver and Internal Medicine Unit, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Wunsch
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Bobinski
- Department of Gastroenterology, Provincial Hospital, Ostroleka, Poland
| | - Michal Mikula
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| |
Collapse
|
46
|
Abstract
The Hippo pathway is an essential tumor suppressor signaling network that coordinates cell proliferation, death, and differentiation in higher eukaryotes. Intriguingly, the core components of the Hippo pathway are conserved from yeast to man, with the yeast analogs of mammalian MST1/2 (fly Hippo), MOB1 (fly Mats), LATS1/2 (fly Warts), and NDR1/2 (fly Tricornered) functioning as essential components of the mitotic exit network (MEN). Here, we update our previous summary of mitotic functions of Hippo core components in Drosophila melanogaster and mammals, with particular emphasis on similarities between the yeast MEN pathway and mitotic Hippo signaling. Mitotic functions of YAP and TAZ, the two main effectors of Hippo signaling, are also discussed.
Collapse
Affiliation(s)
- Alexander Hergovich
- Tumour Suppressor Signalling Networks Laboratory, UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6BT, UK.
| |
Collapse
|
47
|
Phosphorylation-Dependent Regulation of the DNA Damage Response of Adaptor Protein KIBRA in Cancer Cells. Mol Cell Biol 2016; 36:1354-65. [PMID: 26929199 DOI: 10.1128/mcb.01004-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/12/2016] [Indexed: 11/20/2022] Open
Abstract
Multifunctional adaptor proteins encompassing various protein-protein interaction domains play a central role in the DNA damage response pathway. In this report, we show that KIBRA is a physiologically interacting reversible substrate of ataxia telangiectasia mutated (ATM) kinase. We identified the site of phosphorylation in KIBRA as threonine 1006, which is embedded within the serine/threonine (S/T) Q consensus motif, by site-directed mutagenesis, and we further confirmed the same with a phospho-(S/T) Q motif-specific antibody. Results from DNA repair functional assays such as the γ-H2AX assay, pulsed-field gel electrophoresis (PFGE), Comet assay, terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, and clonogenic cell survival assay using stable overexpression clones of wild-type (wt.) KIBRA and active (T1006E) and inactive (T1006A) KIBRA phosphorylation mutants showed that T1006 phosphorylation on KIBRA is essential for optimal DNA double-strand break repair in cancer cells. Further, results from stable retroviral short hairpin RNA-mediated knockdown (KD) clones of KIBRA and KIBRA knockout (KO) model cells generated by a clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 system showed that depleting KIBRA levels compromised the DNA repair functions in cancer cells upon inducing DNA damage. All these phenotypic events were reversed upon reconstitution of KIBRA into cells lacking KIBRA knock-in (KI) model cells. All these results point to the fact that phosphorylated KIBRA might be functioning as a scaffolding protein/adaptor protein facilitating the platform for further recruitment of other DNA damage response factors. In summary, these data demonstrate the imperative functional role of KIBRAper se(KIBRA phosphorylation at T1006 site as a molecular switch that regulates the DNA damage response, possibly via the nonhomologous end joining [NHEJ] pathway), suggesting that KIBRA could be a potential therapeutic target for modulating chemoresistance in cancer cells.
Collapse
|
48
|
Stauffer S, Chen X, Zhang L, Chen Y, Dong J. KIBRA promotes prostate cancer cell proliferation and motility. FEBS J 2016; 283:1800-11. [PMID: 27220053 DOI: 10.1111/febs.13718] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 11/27/2022]
Abstract
KIBRA is a regulator of the Hippo-yes-associated protein (YAP) pathway, which plays a critical role in tumorigenesis. In the present study, we show that KIBRA is a positive regulator in prostate cancer cell proliferation and motility. We found that KIBRA is transcriptionally upregulated in androgen-insensitive LNCaPC4-2 and LNCaP-C81 cells compared to parental androgen-sensitive LNCaP cells. Ectopic expression of KIBRA enhances cell proliferation, migration and invasion in both immortalized and cancerous prostate epithelial cells. Accordingly, knockdown of KIBRA reduces migration, invasion and anchorage-independent growth in LNCaP-C4-2/C81 cells. Moreover, KIBRA expression is induced by androgen signaling and KIBRA is partially required for androgen receptor signaling activation in prostate cancer cells. In line with these findings, we further show that KIBRA is overexpressed in human prostate tumors. Our studies uncover unexpected results and identify KIBRA as a tumor promoter in prostate cancer.
Collapse
Affiliation(s)
- Seth Stauffer
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lin Zhang
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
49
|
Tracy TE, Sohn PD, Minami SS, Wang C, Min SW, Li Y, Zhou Y, Le D, Lo I, Ponnusamy R, Cong X, Schilling B, Ellerby LM, Huganir RL, Gan L. Acetylated Tau Obstructs KIBRA-Mediated Signaling in Synaptic Plasticity and Promotes Tauopathy-Related Memory Loss. Neuron 2016; 90:245-60. [PMID: 27041503 DOI: 10.1016/j.neuron.2016.03.005] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/17/2016] [Accepted: 02/22/2016] [Indexed: 11/27/2022]
Abstract
Tau toxicity has been implicated in the emergence of synaptic dysfunction in Alzheimer's disease (AD), but the mechanism by which tau alters synapse physiology and leads to cognitive decline is unclear. Here we report abnormal acetylation of K274 and K281 on tau, identified in AD brains, promotes memory loss and disrupts synaptic plasticity by reducing postsynaptic KIdney/BRAin (KIBRA) protein, a memory-associated protein. Transgenic mice expressing human tau with lysine-to-glutamine mutations to mimic K274 and K281 acetylation (tauKQ) exhibit AD-related memory deficits and impaired hippocampal long-term potentiation (LTP). TauKQ reduces synaptic KIBRA levels and disrupts activity-induced postsynaptic actin remodeling and AMPA receptor insertion. The LTP deficit was rescued by promoting actin polymerization or by KIBRA expression. In AD patients with dementia, we found enhanced tau acetylation is linked to loss of KIBRA. These findings suggest a novel mechanism by which pathogenic tau causes synaptic dysfunction and cognitive decline in AD pathogenesis.
Collapse
Affiliation(s)
- Tara E Tracy
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Peter Dongmin Sohn
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 91458, USA
| | - S Sakura Minami
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Sang-Won Min
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | - David Le
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA
| | | | - Xin Cong
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA 91458, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 91458, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 91458, USA.
| |
Collapse
|
50
|
Talarowska M, Szemraj J, Kowalczyk M, Gałecki P. Serum KIBRA mRNA and Protein Expression and Cognitive Functions in Depression. Med Sci Monit 2016; 22:152-60. [PMID: 26768155 PMCID: PMC4716708 DOI: 10.12659/msm.895200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Genes participating in synaptic signalling or plasticity in brain regions such as the prefrontal cortex (PFC) and the hippocampus have been implicated in cognition. Recently, a new gene (KIBRA, WWC1) has been added to this group due to its impact on memory performance. Recurrent depressive disorder (rDD) is a multifactorial disease, that one of the typical features is cognitive impairment. The main objective of this study was to perform an analysis of the KIBRA gene on both mRNA and protein levels in patients suffering from rDD and to investigate the relationship between KIBRA expression and cognitive performance. MATERIAL/METHODS The study comprised 236 subjects: patients with rDD (n=131) and healthy subjects (n=105, HS). Cognitive function assessment was based on: Trail Making Test, The Stroop Test, Verbal Fluency Test and Auditory Verbal Learning Test. RESULTS Both mRNA and protein expression levels of KIBRA gene were significantly higher in healthy subjects when compared to rDD. The presented relationship is clear even after taking age, education and sex of the examined subjects into consideration. No statistically significant relationship was found in the experiments between any of the conducted tests and KIBRA gene expression on mRNA level for both the rDD and HS groups. The presented study has limitations related to the fact that patients were being treated with antidepressant. This is relevant due to the fact that some antidepressants may affect mRNA expression. Number of patients and healthy subjects may result in the lack of statistical significance in some cases. CONCLUSIONS 1. The results of our study show decreased expression of the KIBRA gene on both mRNA and protein levels in depression. 2. We did not find any significant relationship between KIBRA gene expression and cognitive functions in case of both the healthy subjects and the patients affected by rDD.
Collapse
Affiliation(s)
- Monika Talarowska
- Department of Adult Psychiatry, Medical University of Łódź, Łódź, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Łódź, Łódź, Poland
| | | | - Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Łódź, Łódź, Poland
| |
Collapse
|