1
|
Liao Y, Muntean BS. KCTD1 regulation of Adenylyl cyclase type 5 adjusts striatal cAMP signaling. Proc Natl Acad Sci U S A 2024; 121:e2406686121. [PMID: 39413138 PMCID: PMC11513970 DOI: 10.1073/pnas.2406686121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024] Open
Abstract
Dopamine transfers information to striatal neurons, and disrupted neurotransmission leads to motor deficits observed in movement disorders. Striatal dopamine converges downstream to Adenylyl Cyclase Type 5 (AC5)-mediated synthesis of cAMP, indicating the essential role of signal transduction in motor physiology. However, the relationship between dopamine decoding and AC5 regulation is unknown. Here, we utilized an unbiased global protein stability screen to identify Potassium Channel Tetramerization Domain 1 (KCTD1) as a key regulator of AC5 level that is mechanistically tied to N-linked glycosylation. We then implemented a CRISPR/SaCas9 approach to eliminate KCTD1 in striatal neurons expressing a Förster resonance energy transfer (FRET)-based cAMP biosensor. 2-photon imaging of striatal neurons in intact circuits uncovered that dopaminergic signaling was substantially compromised in the absence of KCTD1. Finally, knockdown of KCTD1 in genetically defined dorsal striatal neurons significantly altered motor behavior in mice. These results reveal that KCTD1 acts as an essential modifier of dopaminergic signaling by stabilizing striatal AC5.
Collapse
Affiliation(s)
- Yini Liao
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA30912
| | - Brian S. Muntean
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA30912
| |
Collapse
|
2
|
Pinkas DM, Bufton JC, Hunt AE, Manning CE, Richardson W, Bullock AN. A BTB extension and ion-binding domain contribute to the pentameric structure and TFAP2A binding of KCTD1. Structure 2024; 32:1586-1593.e4. [PMID: 39191250 DOI: 10.1016/j.str.2024.07.023] [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: 06/14/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024]
Abstract
KCTD family proteins typically assemble into cullin-RING E3 ligases. KCTD1 is an atypical member that functions instead as a transcriptional repressor. Mutations in KCTD1 cause developmental abnormalities and kidney fibrosis in scalp-ear-nipple syndrome. Here, we present unexpected mechanistic insights from the structure of human KCTD1. Disease-causing mutation P20S maps to an unrecognized extension of the BTB domain that contributes to both its pentameric structure and TFAP2A binding. The C-terminal domain (CTD) shares its fold and pentameric assembly with the GTP cyclohydrolase I feedback regulatory protein (GFRP) despite lacking discernible sequence similarity. Most surprisingly, the KCTD1 CTD establishes a central channel occupied by alternating sodium and iodide ions that restrict TFAP2A dissociation. The elucidation of the structure redefines the KCTD1 BTB domain fold and identifies an unexpected ion-binding site for future study of KCTD1's function in the ectoderm, neural crest, and kidney.
Collapse
Affiliation(s)
- Daniel M Pinkas
- Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK.
| | - Joshua C Bufton
- Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Alice E Hunt
- Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Charlotte E Manning
- Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - William Richardson
- Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Alex N Bullock
- Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK.
| |
Collapse
|
3
|
Balasco N, Ruggiero A, Smaldone G, Pecoraro G, Coppola L, Pirone L, Pedone EM, Esposito L, Berisio R, Vitagliano L. Structural studies of KCTD1 and its disease-causing mutant P20S provide insights into the protein function and misfunction. Int J Biol Macromol 2024; 277:134390. [PMID: 39111466 DOI: 10.1016/j.ijbiomac.2024.134390] [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: 06/19/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/11/2024]
Abstract
Members of the KCTD protein family play key roles in fundamental physio-pathological processes including cancer, neurodevelopmental/neuropsychiatric, and genetic diseases. Here, we report the crystal structure of the KCTD1 P20S mutant, which causes the scalp-ear-nipple syndrome, and molecular dynamics (MD) data on the wild-type protein. Surprisingly, the structure unravels that the N-terminal region, which precedes the BTB domain (preBTB) and bears the disease-associated mutation, adopts a folded polyproline II (PPII) state. The KCTD1 pentamer is characterized by an intricate architecture in which the different subunits mutually exchange domains to generate a closed domain swapping motif. Indeed, the BTB of each chain makes peculiar contacts with the preBTB and the C-terminal domain (CTD) of an adjacent chain. The BTB-preBTB interaction consists of a PPII-PPII recognition motif whereas the BTB-CTD contacts are mediated by an unusual (+/-) helix discontinuous association. The inspection of the protein structure, along with the data emerged from the MD simulations, provides an explanation of the pathogenicity of the P20S mutation and unravels the role of the BTB-preBTB interaction in the insurgence of the disease. Finally, the presence of potassium bound to the central cavity of the CTD pentameric assembly provides insights into the role of KCTD1 in metal homeostasis.
Collapse
Affiliation(s)
- Nicole Balasco
- Institute of Molecular Biology and Pathology, CNR c/o Department Chemistry, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessia Ruggiero
- Institute of Molecular Biology and Pathology, CNR c/o Department Chemistry, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | | | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy
| | - Emilia M Pedone
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy
| | - Luciana Esposito
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy.
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy.
| |
Collapse
|
4
|
Balasco N, Esposito L, Smaldone G, Salvatore M, Vitagliano L. A Comprehensive Analysis of the Structural Recognition between KCTD Proteins and Cullin 3. Int J Mol Sci 2024; 25:1881. [PMID: 38339159 PMCID: PMC10856315 DOI: 10.3390/ijms25031881] [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: 12/22/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
KCTD ((K)potassium Channel Tetramerization Domain-containing) proteins constitute an emerging class of proteins involved in fundamental physio-pathological processes. In these proteins, the BTB domain, which represents the defining element of the family, may have the dual role of promoting oligomerization and favoring functionally important partnerships with different interactors. Here, by exploiting the potential of recently developed methodologies for protein structure prediction, we report a comprehensive analysis of the interactions of all KCTD proteins with their most common partner Cullin 3 (Cul3). The data here presented demonstrate the impressive ability of this approach to discriminate between KCTDs that interact with Cul3 and those that do not. Indeed, reliable and stable models of the complexes were only obtained for the 15 members of the family that are known to interact with Cul3. The generation of three-dimensional models for all KCTD-Cul3 complexes provides interesting clues on the determinants of the structural basis of this partnership as clear structural differences emerged between KCTDs that bind or do not bind Cul3. Finally, the availability of accurate three-dimensional models for KCTD-Cul3 interactions may be valuable for the ad hoc design and development of compounds targeting specific KCTDs that are involved in several common diseases.
Collapse
Affiliation(s)
- Nicole Balasco
- Institute of Molecular Biology and Pathology, CNR c/o Department Chemistry, Sapienza University of Rome, 00185 Rome, Italy
| | - Luciana Esposito
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy;
| | | | | | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, CNR, 80131 Naples, Italy;
| |
Collapse
|
5
|
Liao Y, Sloan DC, Widjaja JH, Muntean BS. KCTD5 Forms Hetero-Oligomeric Complexes with Various Members of the KCTD Protein Family. Int J Mol Sci 2023; 24:14317. [PMID: 37762619 PMCID: PMC10531988 DOI: 10.3390/ijms241814317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Potassium Channel Tetramerization Domain 5 (KCTD5) regulates diverse aspects of physiology, ranging from neuronal signaling to colorectal cancer. A key feature of KCTD5 is its self-assembly into multi-subunit oligomers that seemingly enables participation in an array of protein-protein interactions. KCTD5 has recently been reported to form hetero-oligomeric complexes with two similar KCTDs (KCTD2 and KCTD17). However, it is not known if KCTD5 forms hetero-oligomeric complexes with the remaining KCTD protein family which contains over two dozen members. Here, we demonstrate that KCTD5 interacts with various KCTD proteins when assayed through co-immunoprecipitation in lysed cells. We reinforced this dataset by examining KCTD5 interactions in a live-cell bioluminescence resonance energy transfer (BRET)-based approach. Finally, we developed an IP-luminescence approach to map regions on KCTD5 required for interaction with a selection of KCTD that have established roles in neuronal signaling. We report that different regions on KCTD5 are responsible for uniquely contributing to interactions with other KCTD proteins. While our results help unravel additional interaction partners for KCTD5, they also reveal additional complexities in KCTDs' biology. Moreover, our findings also suggest that KCTD hetero-oligomeric interactions may occur throughout the KCTD family.
Collapse
Affiliation(s)
| | | | | | - Brian S. Muntean
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (Y.L.); (J.H.W.)
| |
Collapse
|
6
|
Di Fiore A, Bellardinelli S, Pirone L, Russo R, Angrisani A, Terriaca G, Bowen M, Bordin F, Besharat ZM, Canettieri G, Fabretti F, Di Gaetano S, Di Marcotullio L, Pedone E, Moretti M, De Smaele E. KCTD1 is a new modulator of the KCASH family of Hedgehog suppressors. Neoplasia 2023; 43:100926. [PMID: 37597490 PMCID: PMC10462845 DOI: 10.1016/j.neo.2023.100926] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/21/2023]
Abstract
The Sonic Hedgehog (Hh) signal transduction pathway plays a critical role in many developmental processes and, when deregulated, may contribute to several cancers, including basal cell carcinoma, medulloblastoma, colorectal, prostate, and pancreatic cancer. In recent years, several Hh inhibitors have been developed, mainly acting on the Smo receptor. However, drug resistance due to Smo mutations or non-canonical Hh pathway activation highlights the need to identify further mechanisms of Hh pathway modulation. Among these, deacetylation of the Hh transcription factor Gli1 by the histone deacetylase HDAC1 increases Hh activity. On the other end, the KCASH family of oncosuppressors binds HDAC1, leading to its ubiquitination and subsequent proteasomal degradation, leaving Gli1 acetylated and not active. It was recently demonstrated that the potassium channel containing protein KCTD15 is able to interact with KCASH2 protein and stabilize it, enhancing its effect on HDAC1 and Hh pathway. KCTD15 and KCTD1 proteins share a high homology and are clustered in a specific KCTD subfamily. We characterize here KCTD1 role on the Hh pathway. Therefore, we demonstrated KCTD1 interaction with KCASH1 and KCASH2 proteins, and its role in their stabilization by reducing their ubiquitination and proteasome-mediated degradation. Consequently, KCTD1 expression reduces HDAC1 protein levels and Hh/Gli1 activity, inhibiting Hh dependent cell proliferation in Hh tumour cells. Furthermore, analysis of expression data on publicly available databases indicates that KCTD1 expression is reduced in Hh dependent MB samples, compared to normal cerebella, suggesting that KCTD1 may represent a new putative target for therapeutic approaches against Hh-dependent tumour.
Collapse
Affiliation(s)
- A Di Fiore
- Department of Experimental Medicine, Sapienza University of Rome, Italy; Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - S Bellardinelli
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - L Pirone
- Institute of Biostructures and Bioimaging, CNR, Naples 80131, Italy
| | - R Russo
- Institute of Biostructures and Bioimaging, CNR, Naples 80131, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli, Caserta, Italy
| | - A Angrisani
- Department of Experimental Medicine, Sapienza University of Rome, Italy; Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - G Terriaca
- Department of Experimental Medicine, Sapienza University of Rome, Italy; Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - M Bowen
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - F Bordin
- Department of Experimental Medicine, Sapienza University of Rome, Italy; Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Z M Besharat
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - G Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - F Fabretti
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - S Di Gaetano
- Institute of Biostructures and Bioimaging, CNR, Naples 80131, Italy
| | - L Di Marcotullio
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - E Pedone
- Institute of Biostructures and Bioimaging, CNR, Naples 80131, Italy
| | - M Moretti
- Department of Experimental Medicine, Sapienza University of Rome, Italy; Neuromed Institute, Pozzilli 86077, Italy
| | - E De Smaele
- Department of Experimental Medicine, Sapienza University of Rome, Italy.
| |
Collapse
|
7
|
Buono L, Iside C, Pecoraro G, De Matteo A, Beneduce G, Penta de Vera d'Aragona R, Parasole R, Mirabelli P, Vitagliano L, Salvatore M, Smaldone G. A Comprehensive Analysis of the Expression Profiles of KCTD Proteins in Acute Lymphoblastic Leukemia: Evidence of Selective Expression of KCTD1 in T-ALL. J Clin Med 2023; 12:jcm12113669. [PMID: 37297863 DOI: 10.3390/jcm12113669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Acute leukemia is the most common pediatric cancer. In most cases, this disease results from the malignant transformation of either the B-cell (B-ALL) or, less frequently, T-cell progenitors (T-ALL). Recently, a marked overexpression of KCTD15, a member of the emerging class of the potassium (K) channel tetramerization domain-containing proteins (KCTDs) has been detected in both patients and continuous cell lines as in vitro model systems. Because there is growing evidence of the key, yet diversified, roles played by KCTDs in cancers, we here report an exhaustive analysis of their expression profiles in both B-ALL and T-ALL patients. Although for most KCTDs, no significant alterations were found in these pathological states, for some members of the family, significant up- and down-regulations were detected in comparison with the values found in healthy subjects in the transcriptome analysis. Among these, particularly relevant is the upregulation of the closely related KCTD1 and KCTD15 in T-ALL patients. Interestingly, KCTD1 is barely expressed in both unaffected controls and B-ALL patients. Therefore, not only does this analysis represent the first study in which the dysregulation of all KCTDs is simultaneously evaluated in specific pathological contexts, but it also provides a promising T-ALL biomarker that could be suitable for clinical applications.
Collapse
Affiliation(s)
- Lorena Buono
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143 Naples, Italy
| | - Concetta Iside
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143 Naples, Italy
| | | | - Antonia De Matteo
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Children's Hospital, AORN, 80122 Naples, Italy
| | - Giuliana Beneduce
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Children's Hospital, AORN, 80122 Naples, Italy
| | | | - Rosanna Parasole
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Children's Hospital, AORN, 80122 Naples, Italy
| | - Peppino Mirabelli
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Children's Hospital, AORN, 80122 Naples, Italy
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, C.N.R., 80134 Napoli, Italy
| | | | | |
Collapse
|
8
|
Yumimoto K, Sugiyama S, Motomura S, Takahashi D, Nakayama KI. Molecular evolution of Keap1 was essential for adaptation of vertebrates to terrestrial life. SCIENCE ADVANCES 2023; 9:eadg2379. [PMID: 37205751 DOI: 10.1126/sciadv.adg2379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Reactive oxygen species (ROS) posed a risk for the transition of vertebrates from aquatic to terrestrial life. How ancestral organisms adapted to such ROS exposure has remained a mystery. Here, we show that attenuation of the activity of the ubiquitin ligase CRL3Keap1 for the transcription factor Nrf2 during evolution was key to development of an efficient response to ROS exposure. The Keap1 gene was duplicated in fish to give rise to Keap1A and the only remaining mammalian paralog Keap1B, the latter of which shows a lower affinity for Cul3 and contributes to robust Nrf2 induction in response to ROS exposure. Mutation of mammalian Keap1 to resemble zebrafish Keap1A resulted in an attenuated Nrf2 response, and most knock-in mice expressing such a Keap1 mutant died on exposure as neonates to sunlight-level ultraviolet radiation. Our results suggest that molecular evolution of Keap1 was essential for adaptation to terrestrial life.
Collapse
Affiliation(s)
- Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Saori Motomura
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Daisuke Takahashi
- Department of Protein Structure, Function, and Design, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
9
|
Wang W, Su L, Meng L, He J, Tan C, Yi D, Cheng D, Zhang H, Lu G, Du J, Lin G, Zhang Q, Tu C, Tan YQ. Biallelic variants in KCTD19 associated with male factor infertility and oligoasthenoteratozoospermia. Hum Reprod 2023:7165695. [PMID: 37192818 DOI: 10.1093/humrep/dead095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 04/14/2023] [Indexed: 05/18/2023] Open
Abstract
STUDY QUESTION Can whole-exome sequencing (WES) reveal new genetic factors responsible for male infertility characterized by oligozoospermia? SUMMARY ANSWER We identified biallelic missense variants in the Potassium Channel Tetramerization Domain Containing 19 gene (KCTD19) and confirmed it to be a novel pathogenic gene for male infertility. WHAT IS KNOWN ALREADY KCTD19 is a key transcriptional regulator that plays an indispensable role in male fertility by regulating meiotic progression. Kctd19 gene-disrupted male mice exhibit infertility due to meiotic arrest. STUDY DESIGN, SIZE, DURATION We recruited a cohort of 536 individuals with idiopathic oligozoospermia from 2014 to 2022 and focused on five infertile males from three unrelated families. Semen analysis data and ICSI outcomes were collected. WES and homozygosity mapping were performed to identify potential pathogenic variants. The pathogenicity of the identified variants was investigated in silico and in vitro. PARTICIPANTS/MATERIALS, SETTING, METHODS Male patients diagnosed with primary infertility were recruited from the Reproductive and Genetic Hospital of CITIC-Xiangya. Genomic DNA extracted from affected individuals was used for WES and Sanger sequencing. Sperm phenotype, sperm nuclear maturity, chromosome aneuploidy, and sperm ultrastructure were assessed using hematoxylin and eosin staining and toluidine blue staining, FISH and transmission electron microscopy. The functional effects of the identified variants in HEK293T cells were investigated via western blotting and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE We identified three homozygous missense variants (NM_001100915, c.G628A:p.E210K, c.C893T:p.P298L, and c.G2309A:p.G770D) in KCTD19 in five infertile males from three unrelated families. Abnormal morphology of the sperm heads with immature nuclei and/or nuclear aneuploidy were frequently observed in individuals with biallelic KCTD19 variants, and ICSI was unable to rescue these deficiencies. These variants reduced the abundance of KCTD19 due to increased ubiquitination and impaired its nuclear colocalization with its functional partner, zinc finger protein 541 (ZFP541), in HEK293T cells. LIMITATIONS, REASONS FOR CAUTION The exact pathogenic mechanism remains unclear, and warrants further studies using knock-in mice that mimic the missense mutations found in individuals with biallelic KCTD19 variants. WIDER IMPLICATIONS OF THE FINDINGS Our study is the first to report a likely causal relationship between KCTD19 deficiency and male infertility, confirming the critical role of KCTD19 in human reproduction. Additionally, this study provided evidence for the poor ICSI clinical outcomes in individuals with biallelic KCTD19 variants, which may guide clinical treatment strategies. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Key Research and Developmental Program of China (2022YFC2702604 to Y.-Q.T.), the National Natural Science Foundation of China (81971447 and 82171608 to Y.-Q.T., 82101961 to C.T.), a key grant from the Prevention and Treatment of Birth Defects from Hunan Province (2019SK1012 to Y.-Q.T.), a Hunan Provincial Grant for Innovative Province Construction (2019SK4012), and the China Postdoctoral Science Foundation (2022M721124 to W.W.). The authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Weili Wang
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Lilan Su
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
| | - Lanlan Meng
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Jiaxin He
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
| | - Chen Tan
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
| | - Duo Yi
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Dehua Cheng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Huan Zhang
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Guangxiu Lu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Juan Du
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Ge Lin
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Qianjun Zhang
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- College of Life Science, Hunan Normal University, Changsha, China
| | - Chaofeng Tu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yue-Qiu Tan
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- College of Life Science, Hunan Normal University, Changsha, China
| |
Collapse
|
10
|
Sharma J, Mulherkar S, Chen UI, Xiong Y, Bajaj L, Cho BK, Goo YA, Leung HCE, Tolias KF, Sardiello M. Calpain activity is negatively regulated by a KCTD7-Cullin-3 complex via non-degradative ubiquitination. Cell Discov 2023; 9:32. [PMID: 36964131 PMCID: PMC10038992 DOI: 10.1038/s41421-023-00533-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/24/2023] [Indexed: 03/26/2023] Open
Abstract
Calpains are a class of non-lysosomal cysteine proteases that exert their regulatory functions via limited proteolysis of their substrates. Similar to the lysosomal and proteasomal systems, calpain dysregulation is implicated in the pathogenesis of neurodegenerative disease and cancer. Despite intensive efforts placed on the identification of mechanisms that regulate calpains, however, calpain protein modifications that regulate calpain activity are incompletely understood. Here we show that calpains are regulated by KCTD7, a cytosolic protein of previously uncharacterized function whose pathogenic mutations result in epilepsy, progressive ataxia, and severe neurocognitive deterioration. We show that KCTD7 works in complex with Cullin-3 and Rbx1 to execute atypical, non-degradative ubiquitination of calpains at specific sites (K398 of calpain 1, and K280 and K674 of calpain 2). Experiments based on single-lysine mutants of ubiquitin determined that KCTD7 mediates ubiquitination of calpain 1 via K6-, K27-, K29-, and K63-linked chains, whereas it uses K6-mediated ubiquitination to modify calpain 2. Loss of KCTD7-mediated ubiquitination of calpains led to calpain hyperactivation, aberrant cleavage of downstream targets, and caspase-3 activation. CRISPR/Cas9-mediated knockout of Kctd7 in mice phenotypically recapitulated human KCTD7 deficiency and resulted in calpain hyperactivation, behavioral impairments, and neurodegeneration. These phenotypes were largely prevented by pharmacological inhibition of calpains, thus demonstrating a major role of calpain dysregulation in KCTD7-associated disease. Finally, we determined that Cullin-3-KCTD7 mediates ubiquitination of all ubiquitous calpains. These results unveil a novel mechanism and potential target to restrain calpain activity in human disease and shed light on the molecular pathogenesis of KCTD7-associated disease.
Collapse
Affiliation(s)
- Jaiprakash Sharma
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, Genetics and Genomic Medicine, Saint Louis, MO, USA.
| | - Shalaka Mulherkar
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, Genetics and Genomic Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Uan-I Chen
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yan Xiong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, Genetics and Genomic Medicine, Saint Louis, MO, USA
| | - Lakshya Bajaj
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Byoung-Kyu Cho
- Mass Spectrometry Technology Access Center at the McDonnell Genome Institute, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Young Ah Goo
- Mass Spectrometry Technology Access Center at the McDonnell Genome Institute, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Hon-Chiu Eastwood Leung
- Departments of Medicine, Pediatrics, and Molecular and Cellular Biology, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Department of Pediatrics, Washington University in St. Louis, School of Medicine, Genetics and Genomic Medicine, Saint Louis, MO, USA.
| |
Collapse
|
11
|
Smaldone G, Pecoraro G, Pane K, Franzese M, Ruggiero A, Vitagliano L, Salvatore M. The Oncosuppressive Properties of KCTD1: Its Role in Cell Growth and Mobility. BIOLOGY 2023; 12:biology12030481. [PMID: 36979172 PMCID: PMC10045846 DOI: 10.3390/biology12030481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
The KCTD protein family is traditionally regarded as proteins that play key roles in neurological physiopathology. However, new studies are increasingly demonstrating their involvement in many other biological processes, including cancers. This is particularly evident for KCTD proteins not involved in protein ubiquitination and degradation, such as KCTD1. We explored the role of KCTD1 in colorectal cancer by knocking down this protein in the human colon adenocarcinoma cell line, SW480. We re-assessed its ability to downregulate β-catenin, a central actor in the WNT/β-catenin signalling pathway. Interestingly, opposite effects are observed when the protein is upregulated in CACO2 colorectal cancer cells. Moreover, interrogation of the TCGA database indicates that KCTD1 downregulation is associated with β-catenin overexpression in colorectal cancer patients. Indeed, knocking down KCTD1 in SW480 cells led to a significant increase in their motility and stemness, two important tumorigenesis traits, suggesting an oncosuppressor role for KCTD1. It is worth noting that similar effects are induced on colorectal cancer cells by the misregulation of KCTD12, a protein that is distantly related to KCTD1. The presented results further expand the spectrum of KCTD1 involvement in apparently unrelated physiopathological processes. The similar effects produced on colorectal cancer cell lines by KCTD1 and KCTD12 suggest novel, previously unreported analogous activities among members of the KCTD protein family.
Collapse
Affiliation(s)
| | | | - Katia Pane
- IRCCS SYNLAB SDN, Via E. Gianturco 113, 80143 Naples, Italy
| | | | - Alessia Ruggiero
- Institute of Biostructures and Bioimaging, C.N.R., 80134 Napoli, Italy
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, C.N.R., 80134 Napoli, Italy
| | | |
Collapse
|
12
|
Multiple potassium channel tetramerization domain (KCTD) family members interact with Gβγ, with effects on cAMP signaling. J Biol Chem 2023; 299:102924. [PMID: 36736897 PMCID: PMC9976452 DOI: 10.1016/j.jbc.2023.102924] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) initiate an array of intracellular signaling programs by activating heterotrimeric G proteins (Gα and Gβγ subunits). Therefore, G protein modifiers are well positioned to shape GPCR pharmacology. A few members of the potassium channel tetramerization domain (KCTD) protein family have been found to adjust G protein signaling through interaction with Gβγ. However, comprehensive details on the KCTD interaction with Gβγ remain unresolved. Here, we report that nearly all the 25 KCTD proteins interact with Gβγ. In this study, we screened Gβγ interaction capacity across the entire KCTD family using two parallel approaches. In a live cell bioluminescence resonance energy transfer-based assay, we find that roughly half of KCTD proteins interact with Gβγ in an agonist-induced fashion, whereas all KCTD proteins except two were found to interact through coimmunoprecipitation. We observed that the interaction was dependent on an amino acid hot spot in the C terminus of KCTD2, KCTD5, and KCTD17. While KCTD2 and KCTD5 require both the Bric-à-brac, Tramtrack, Broad complex domain and C-terminal regions for Gβγ interaction, we uncovered that the KCTD17 C terminus is sufficient for Gβγ interaction. Finally, we demonstrated the functional consequence of the KCTD-Gβγ interaction by examining sensitization of the adenylyl cyclase-cAMP pathway in live cells. We found that Gβγ-mediated sensitization of adenylyl cyclase 5 was blunted by KCTD. We conclude that the KCTD family broadly engages Gβγ to shape GPCR signal transmission.
Collapse
|
13
|
AlphaFold-Predicted Structures of KCTD Proteins Unravel Previously Undetected Relationships among the Members of the Family. Biomolecules 2021; 11:biom11121862. [PMID: 34944504 PMCID: PMC8699099 DOI: 10.3390/biom11121862] [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: 11/09/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
One of the most striking features of KCTD proteins is their involvement in apparently unrelated yet fundamental physio-pathological processes. Unfortunately, comprehensive structure–function relationships for this protein family have been hampered by the scarcity of the structural data available. This scenario is rapidly changing due to the release of the protein three-dimensional models predicted by AlphaFold (AF). Here, we exploited the structural information contained in the AF database to gain insights into the relationships among the members of the KCTD family with the aim of facilitating the definition of the structural and molecular basis of key roles that these proteins play in many biological processes. The most important finding that emerged from this investigation is the discovery that, in addition to the BTB domain, the vast majority of these proteins also share a structurally similar domain in the C-terminal region despite the absence of general sequence similarities detectable in this region. Using this domain as reference, we generated a novel and comprehensive structure-based pseudo-phylogenetic tree that unraveled previously undetected similarities among the protein family. In particular, we generated a new clustering of the KCTD proteins that will represent a solid ground for interpreting their many functions.
Collapse
|
14
|
Márquez-Cantudo L, Ramos A, Coderch C, de Pascual-Teresa B. Proteasomal Degradation of Zn-Dependent Hdacs: The E3-Ligases Implicated and the Designed Protacs That Enable Degradation. Molecules 2021; 26:molecules26185606. [PMID: 34577077 PMCID: PMC8467390 DOI: 10.3390/molecules26185606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/21/2022] Open
Abstract
Protein degradation by the Ubiquitin-Proteasome System is one of the main mechanisms of the regulation of cellular proteostasis, and the E3 ligases are the key effectors for the protein recognition and degradation. Many E3 ligases have key roles in cell cycle regulation, acting as checkpoints and checkpoint regulators. One of the many important proteins involved in the regulation of the cell cycle are the members of the Histone Deacetylase (HDAC) family. The importance of zinc dependent HDACs in the regulation of chromatin packing and, therefore, gene expression, has made them targets for the design and synthesis of HDAC inhibitors. However, achieving potency and selectivity has proven to be a challenge due to the homology between the zinc dependent HDACs. PROteolysis TArgeting Chimaera (PROTAC) design has been demonstrated to be a useful strategy to inhibit and selectively degrade protein targets. In this review, we attempt to summarize the E3 ligases that naturally ubiquitinate HDACs, analyze their structure, and list the known ligands that can bind to these E3 ligases and be used for PROTAC design, as well as the already described HDAC-targeted PROTACs.
Collapse
|
15
|
Smaldone G, Coppola L, Pane K, Franzese M, Beneduce G, Parasole R, Menna G, Vitagliano L, Salvatore M, Mirabelli P. KCTD15 deregulation is associated with alterations of the NF-κB signaling in both pathological and physiological model systems. Sci Rep 2021; 11:18237. [PMID: 34521919 PMCID: PMC8440651 DOI: 10.1038/s41598-021-97775-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Like other KCTD proteins, KCTD15 is involved in important albeit distinct biological processes as cancer, neural crest formation, and obesity. Here, we characterized the role of KCTD15 in different physiological/pathological states to gain insights into its diversified function(s). The silencing of KCTD15 in MLL-rearranged leukemia models induced attenuation of the NF-κB pathway associated with a downregulation of pIKK-β and pIKB-α. Conversely, the activation of peripheral blood T cells upon PMA/ionomycin stimulation remarkably upregulated KCTD15 and, simultaneously, pIKK-β and pIKB-α. Moreover, a significant upregulation of KCTD15 was also observed in CD34 hematopoietic stem/progenitor cells where the NF-κB pathway is physiologically activated. The association between KCTD15 upregulation and increased NF-κB signaling was confirmed by luciferase assay as well as KCTD15 and IKK-β proximity ligation and immunoprecipitation experiments. The observed upregulation of IKK-β by KCTD15 provides a novel and intriguing interpretative key for understanding the protein function in a wide class of physiological/pathological conditions ranging from neuronal development to cancer and obesity/diabetes.
Collapse
Affiliation(s)
| | - Luigi Coppola
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - Katia Pane
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | | | - Giuliana Beneduce
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Hospital, 80129, Naples, Italy
| | - Rosanna Parasole
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Hospital, 80129, Naples, Italy
| | - Giuseppe Menna
- Department of Pediatric Hemato-Oncology, Santobono-Pausilipon Hospital, 80129, Naples, Italy
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, C.N.R., Via Mezzocannone n.16, 80134, Naples, Italy.
| | | | | |
Collapse
|
16
|
Chen W, Su J, Cai S, Shi C. Cullin3 aggravates the inflammatory response of periodontal ligament stem cells via regulation of SHH signaling and Nrf2. Bioengineered 2021; 12:3089-3100. [PMID: 34193016 PMCID: PMC8806625 DOI: 10.1080/21655979.2021.1943603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It is found that the activation of Sonic Hedgehog (SHH) signaling pathway is related to the degree of inflammation in patients suffering from periodontitis. Cullin3 (CUL3), an important ubiquitin ligase, can control SHH signaling. In this study, we were dedicated to clarify the roles of SHH and CUL3 in P. gingivalis-LPS (Pg-LPS)-treated periodontal ligament stem cells (PDLSCs). In this study, cell viability was detected using cell counting kit-8 (CCK-8). The inflammatory cytokines of PDLSCs were estimated by enzyme-linked immunosorbent assay (ELISA). With the application of western blots, the protein levels of SHH, Gli1 and NF-E2-related factor 2 (Nrf2) were determined. Alkaline phosphatase staining and Alizarin red staining were performed to evaluate the differentiation and mineralization capabilities of PDLSCs. The apoptotic cells were screened using TUNEL staining. The results showed that Pg-LPS inhibited cell viability and triggered inflammation of PDLSCs. Overexpression of CUL3 weakened the differentiation and mineralization capabilities of PDLSCs. Moreover, CUL3 overexpression aggravated inflammation and cell apoptosis induced by Pg-LPS. It is worth noting that although the protein levels of SHH, Gli1 and Nrf2 were elevated in PDLSCs treated with Pg-LPS, overexpression of CUL3 decreased the expressions of Gli1 and Nrf2. Overall, SHH/Gli1 and Nrf2 were involved in the inflammation and cell apoptosis of PDLSCs, which was dominated by CUL3.
Collapse
Affiliation(s)
- Wanhong Chen
- Department of Stomatology, Quanzhou First Hospital, Quanzhou, Fujian, China
| | - Jiangling Su
- Department of Stomatology, Quanzhou First Hospital, Quanzhou, Fujian, China
| | - Shixiong Cai
- Department of Stomatology, Quanzhou First Hospital, Quanzhou, Fujian, China
| | - Chun Shi
- Department of Endodontics and Periodontics, School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
17
|
Angrisani A, Di Fiore A, De Smaele E, Moretti M. The emerging role of the KCTD proteins in cancer. Cell Commun Signal 2021; 19:56. [PMID: 34001146 PMCID: PMC8127222 DOI: 10.1186/s12964-021-00737-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/05/2021] [Indexed: 12/24/2022] Open
Abstract
The human family of Potassium (K+) Channel Tetramerization Domain (KCTD) proteins counts 25 members, and a significant number of them are still only partially characterized. While some of the KCTDs have been linked to neurological disorders or obesity, a growing tally of KCTDs are being associated with cancer hallmarks or involved in the modulation of specific oncogenic pathways. Indeed, the potential relevance of the variegate KCTD family in cancer warrants an updated picture of the current knowledge and highlights the need for further research on KCTD members as either putative therapeutic targets, or diagnostic/prognostic markers. Homology between family members, capability to participate in ubiquitination and degradation of different protein targets, ability to heterodimerize between members, role played in the main signalling pathways involved in development and cancer, are all factors that need to be considered in the search for new key players in tumorigenesis. In this review we summarize the recent published evidence on KCTD members' involvement in cancer. Furthermore, by integrating this information with data extrapolated from public databases that suggest new potential associations with cancers, we hypothesize that the number of KCTD family members involved in tumorigenesis (either as positive or negative modulator) may be bigger than so far demonstrated. Video abstract.
Collapse
Affiliation(s)
| | - Annamaria Di Fiore
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Marta Moretti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
18
|
KCTD19 and its associated protein ZFP541 are independently essential for meiosis in male mice. PLoS Genet 2021; 17:e1009412. [PMID: 33961623 PMCID: PMC8104389 DOI: 10.1371/journal.pgen.1009412] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/05/2021] [Indexed: 12/24/2022] Open
Abstract
Meiosis is a cell division process with complex chromosome events where various molecules must work in tandem. To find meiosis-related genes, we screened evolutionarily conserved and reproductive tract-enriched genes using the CRISPR/Cas9 system and identified potassium channel tetramerization domain containing 19 (Kctd19) as an essential factor for meiosis. In prophase I, Kctd19 deficiency did not affect synapsis or the DNA damage response, and chiasma structures were also observed in metaphase I spermatocytes of Kctd19 KO mice. However, spermatocytes underwent apoptotic elimination during the metaphase-anaphase transition. We were able to rescue the Kctd19 KO phenotype with an epitope-tagged Kctd19 transgene. By immunoprecipitation-mass spectrometry, we confirmed the association of KCTD19 with zinc finger protein 541 (ZFP541) and histone deacetylase 1 (HDAC1). Phenotyping of Zfp541 KO spermatocytes demonstrated XY chromosome asynapsis and recurrent DNA damage in the late pachytene stage, leading to apoptosis. In summary, our study reveals that KCTD19 associates with ZFP541 and HDAC1, and that both KCTD19 and ZFP541 are essential for meiosis in male mice. Meiosis is a fundamental process that consists of one round of genomic DNA replication and two rounds of chromosome segregation, producing four haploid cells. To properly distribute their genetic material, cells need to undergo complex chromosome events such as a physical linkage of homologous chromosomes (termed synapsis) and meiotic recombination. The molecules involved in these events have not been fully characterized yet, especially in mammals. Using a CRISPR/Cas9-screening system, we identified the potassium channel tetramerization domain containing 19 (Kctd19) as an essential factor for meiosis in male mice. Further, we confirmed the association of KCTD19 with zinc finger protein 541 (ZFP541) and histone deacetylase 1 (HDAC1). By observing meiosis of Zfp541 knockout germ cells, we found that Zfp541 was also essential for meiosis. These results show that the KCTD19/ZFP541 complex plays a critical role and is indispensable for male meiosis and fertility.
Collapse
|
19
|
Blondelle J, Biju A, Lange S. The Role of Cullin-RING Ligases in Striated Muscle Development, Function, and Disease. Int J Mol Sci 2020; 21:E7936. [PMID: 33114658 PMCID: PMC7672578 DOI: 10.3390/ijms21217936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
The well-orchestrated turnover of proteins in cross-striated muscles is one of the fundamental processes required for muscle cell function and survival. Dysfunction of the intricate protein degradation machinery is often associated with development of cardiac and skeletal muscle myopathies. Most muscle proteins are degraded by the ubiquitin-proteasome system (UPS). The UPS involves a number of enzymes, including E3-ligases, which tightly control which protein substrates are marked for degradation by the proteasome. Recent data reveal that E3-ligases of the cullin family play more diverse and crucial roles in cross striated muscles than previously anticipated. This review highlights some of the findings on the multifaceted functions of cullin-RING E3-ligases, their substrate adapters, muscle protein substrates, and regulatory proteins, such as the Cop9 signalosome, for the development of cross striated muscles, and their roles in the etiology of myopathies.
Collapse
Affiliation(s)
- Jordan Blondelle
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Andrea Biju
- Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Stephan Lange
- Department of Medicine, University of California, La Jolla, CA 92093, USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
20
|
Smaldone G, Coppola L, Incoronato M, Parasole R, Ripaldi M, Vitagliano L, Mirabelli P, Salvatore M. KCTD15 Protein Expression in Peripheral Blood and Acute Myeloid Leukemia. Diagnostics (Basel) 2020; 10:diagnostics10060371. [PMID: 32512747 PMCID: PMC7345863 DOI: 10.3390/diagnostics10060371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 12/24/2022] Open
Abstract
Leukocytes are major cellular components of the inflammatory and immune response systems. After their generation in the bone marrow from hematopoietic stem cells, they maturate as granulocytes (neutrophils, eosinophils, and basophils), monocytes, and lymphocytes. The abnormal accumulation and proliferation of immature blood cells (blasts) lead to severe and widespread diseases such as leukemia. We have recently shown that KCTD15, a member of the potassium channel tetramerization domain containing protein family (KCTD), is remarkably upregulated in leukemic B-cells. Here, we extend our investigation by monitoring the KCTD15 expression levels in circulating lymphocytes, monocytes, and granulocytes, as well as in leukemia cells. Significant differences in the expression level of KCTD15 were detected in normal lymphocytes, monocytes, and granulocytes. Interestingly, we also found overexpression of the protein following leukemic transformation in the case of myeloid cell lineage. Indeed, KCTD15 was found to be upregulated in K562 and NB4 cells, as well as in HL-60 cell lines. This in vitro finding was corroborated by the analysis of KCTD15 mRNA of acute myeloid leukemia (AML) patients reported in the Microarray Innovations in Leukemia (MILE) dataset. Collectively, the present data open interesting perspectives for understanding the maturation process of leukocytes and for the diagnosis/therapy of acute leukemias.
Collapse
Affiliation(s)
- Giovanni Smaldone
- IRCCS SDN, Napoli, Via E. Gianturco 113, 80143 Naples, Italy; (G.S.); (L.C.); (M.I.); (M.S.)
| | - Luigi Coppola
- IRCCS SDN, Napoli, Via E. Gianturco 113, 80143 Naples, Italy; (G.S.); (L.C.); (M.I.); (M.S.)
| | - Mariarosaria Incoronato
- IRCCS SDN, Napoli, Via E. Gianturco 113, 80143 Naples, Italy; (G.S.); (L.C.); (M.I.); (M.S.)
| | - Rosanna Parasole
- Department of Pediatric Hematology-Oncology, Santobono-Pausilipon Hospital, 80129 Naples, Italy; (R.P.); (M.R.)
| | - Mimmo Ripaldi
- Department of Pediatric Hematology-Oncology, Santobono-Pausilipon Hospital, 80129 Naples, Italy; (R.P.); (M.R.)
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, C.N.R., 80134 Napoli, Italy
- Correspondence: (L.V.); (P.M.)
| | - Peppino Mirabelli
- IRCCS SDN, Napoli, Via E. Gianturco 113, 80143 Naples, Italy; (G.S.); (L.C.); (M.I.); (M.S.)
- Correspondence: (L.V.); (P.M.)
| | - Marco Salvatore
- IRCCS SDN, Napoli, Via E. Gianturco 113, 80143 Naples, Italy; (G.S.); (L.C.); (M.I.); (M.S.)
| |
Collapse
|
21
|
KCTD15 is overexpressed in human childhood B-cell acute lymphoid leukemia. Sci Rep 2019; 9:20108. [PMID: 31882877 PMCID: PMC6934626 DOI: 10.1038/s41598-019-56701-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022] Open
Abstract
Leukemic cells originate from the malignant transformation of undifferentiated myeloid/lymphoid hematopoietic progenitors normally residing in bone marrow. As the precise molecular mechanisms underlying this heterogeneous disease are yet to be disclosed, the identification and the validation of novel actors in leukemia is of extreme importance. Here, we show that KCTD15, a member of the emerging class of KCTD ((K)potassium Channel Tetramerization Domain containing) proteins, is strongly upregulated in patients affected by B-cell type acute lymphoblastic leukemia (B-ALL) and in continuous cell lines (RS4;11, REH, TOM-1, SEM) derived from this form of childhood leukemia. Interestingly, KCTD15 downregulation induces apoptosis and cell death suggesting that it has a role in cellular homeostasis and proliferation. In addition, stimulation of normal lymphocytes with the pokeweed mitogen leads to increased KCTD15 levels in a fashion comparable to those observed in proliferating leukemic cells. In this way, the role of KCTD15 is likely not confined to the B-ALL pathological state and extends to activation and proliferation of normal lymphocytes. Collectively, data here presented indicate that KCTD15 is an important and hitherto unidentified player in childhood lymphoid leukemia, and its study could open a new scenario for the identification of altered and still unknown molecular pathways in leukemia.
Collapse
|
22
|
Spiombi E, Angrisani A, Fonte S, De Feudis G, Fabretti F, Cucchi D, Izzo M, Infante P, Miele E, Po A, Di Magno L, Magliozzi R, Guardavaccaro D, Maroder M, Canettieri G, Giannini G, Ferretti E, Gulino A, Di Marcotullio L, Moretti M, De Smaele E. KCTD15 inhibits the Hedgehog pathway in Medulloblastoma cells by increasing protein levels of the oncosuppressor KCASH2. Oncogenesis 2019; 8:64. [PMID: 31685809 PMCID: PMC6828672 DOI: 10.1038/s41389-019-0175-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 12/17/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant childhood brain tumor. About 30% of all MBs belong to the I molecular subgroup, characterized by constitutive activation of the Sonic Hedgehog (Hh) pathway. The Hh pathway is involved in several fundamental processes during embryogenesis and in adult life and its deregulation may lead to cerebellar tumorigenesis. Indeed, Hh activity must be maintained via a complex network of activating and repressor signals. One of these repressor signals is KCASH2, belonging to the KCASH family of protein, which acts as negative regulators of the Hedgehog signaling pathway during cerebellar development and differentiation. KCASH2 leads HDAC1 to degradation, allowing hyperacetylation and inhibition of transcriptional activity of Gli1, the main effector of the Hh pathway. In turn, the KCASH2 loss leads to persistent Hh activity and eventually tumorigenesis. In order to better characterize the physiologic role and modulation mechanisms of KCASH2, we have searched through a proteomic approach for new KCASH2 interactors, identifying Potassium Channel Tetramerization Domain Containing 15 (KCTD15). KCTD15 is able to directly interact with KCASH2, through its BTB/POZ domain. This interaction leads to increase KCASH2 stability which implies a reduction of the Hh pathway activity and a reduction of Hh-dependent MB cells proliferation. Here we report the identification of KCTD15 as a novel player in the complex network of regulatory proteins, which modulate Hh pathway, this could be a promising new target for therapeutic approach against MB.
Collapse
Affiliation(s)
- Eleonora Spiombi
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, 20090, Segrate, Milan, Italy
| | - Annapaola Angrisani
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Simone Fonte
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Giuseppina De Feudis
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Department of Experimental Oncology, European Institute of Oncology, 20139, Milan, Italy
| | - Francesca Fabretti
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Danilo Cucchi
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Barts Cancer Institute, Queen Mary University of London, Centre for Molecular Oncology, John Vane Science Center, London, EC1M 6BQ, UK
| | - Mariapaola Izzo
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | - Paola Infante
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | - Evelina Miele
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Agnese Po
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Laura Di Magno
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | | | | | - Marella Maroder
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, 00161, Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, 00161, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University, 00161, Rome, Italy
| | - Alberto Gulino
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, 00161, Rome, Italy
| | - Marta Moretti
- Department of Experimental Medicine, Sapienza University, 00161, Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University, 00161, Rome, Italy.
| |
Collapse
|
23
|
Targeted protein degradation: expanding the toolbox. Nat Rev Drug Discov 2019; 18:949-963. [PMID: 31666732 DOI: 10.1038/s41573-019-0047-y] [Citation(s) in RCA: 581] [Impact Index Per Article: 96.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2019] [Indexed: 12/19/2022]
Abstract
Proteolysis-targeting chimeras (PROTACs) and related molecules that induce targeted protein degradation by the ubiquitin-proteasome system represent a new therapeutic modality and are the focus of great interest, owing to potential advantages over traditional occupancy-based inhibitors with respect to dosing, side effects, drug resistance and modulating 'undruggable' targets. However, the technology is still maturing, and the design elements for successful PROTAC-based drugs are currently being elucidated. Importantly, fewer than 10 of the more than 600 E3 ubiquitin ligases have so far been exploited for targeted protein degradation, and expansion of knowledge in this area is a key opportunity. Here, we briefly discuss lessons learned about targeted protein degradation in chemical biology and drug discovery and systematically review the expression profile, domain architecture and chemical tractability of human E3 ligases that could expand the toolbox for PROTAC discovery.
Collapse
|
24
|
Pirone L, Smaldone G, Spinelli R, Barberisi M, Beguinot F, Vitagliano L, Miele C, Di Gaetano S, Raciti GA, Pedone E. KCTD1: A novel modulator of adipogenesis through the interaction with the transcription factor AP2α. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:158514. [PMID: 31465887 DOI: 10.1016/j.bbalip.2019.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/09/2019] [Accepted: 08/22/2019] [Indexed: 01/23/2023]
Abstract
Adipogenesis has an important role in regulating energy balance, tissue homeostasis and disease pathogenesis. 3T3-L1 preadipocytes have been widely used as an in vitro model for studying adipocyte differentiation. We here show that KCTD1, a member of the potassium channel containing tetramerization domain proteins, plays an active role in adipogenesis. In particular, we show KCTD1 expression 3T3-L1 cells increases upon adipogenesis induction. Treatment of 3T3-L1 preadipocytes with Kctd1-specific siRNA inhibited the differentiation, as indicated by reduction of expression of the specific adipogenic markers C/ebpα, Pparγ2, Glut4, and Adiponectin. Moreover, we also show that the protein physically interacts with the transcription factor AP2α, a known inhibitor of adipogenesis, both in vitro and in cells. Interestingly, our data indicate that KCTD1 promotes adipogenesis through the interaction with AP2α and by removing it from the nucleus. Collectively, these findings disclose a novel role for KCTD1 and pave the way for novel strategies aimed at modulating adipogenesis.
Collapse
Affiliation(s)
- Luciano Pirone
- Istituto di Biostrutture e Bioimmagini, CNR, Napoli, Italy
| | | | - Rosa Spinelli
- URT "Genomica del Diabete", Istituto per l'Endocrinologia e l'Oncologia Sperimentale "Gaetano Salvatore", CNR, Napoli, Italy; Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Italy
| | - Manlio Barberisi
- Dipartimento Scienze Anastesiologiche, Chirurgiche E Dell'emergenza, Università Della Campania-Luigi Vanvitelli, Caserta, Italy
| | - Francesco Beguinot
- URT "Genomica del Diabete", Istituto per l'Endocrinologia e l'Oncologia Sperimentale "Gaetano Salvatore", CNR, Napoli, Italy; Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Italy
| | | | - Claudia Miele
- URT "Genomica del Diabete", Istituto per l'Endocrinologia e l'Oncologia Sperimentale "Gaetano Salvatore", CNR, Napoli, Italy; Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Italy
| | | | - Gregory Alexander Raciti
- URT "Genomica del Diabete", Istituto per l'Endocrinologia e l'Oncologia Sperimentale "Gaetano Salvatore", CNR, Napoli, Italy; Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Italy
| | - Emilia Pedone
- Istituto di Biostrutture e Bioimmagini, CNR, Napoli, Italy.
| |
Collapse
|
25
|
The Structural Versatility of the BTB Domains of KCTD Proteins and Their Recognition of the GABA B Receptor. Biomolecules 2019; 9:biom9080323. [PMID: 31370201 PMCID: PMC6722564 DOI: 10.3390/biom9080323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
Several recent investigations have demonstrated that members of the KCTD (Potassium Channel Tetramerization Domain) protein family are involved in fundamental processes. However, the paucity of structural data available on these proteins has frequently prevented the definition of their biochemical role(s). Fortunately, this scenario is rapidly changing as, in very recent years, several crystallographic structures have been reported. Although these investigations have provided very important insights into the function of KCTDs, they have also raised some puzzling issues. One is related to the observation that the BTB (broad-complex, tramtrack, and bric-à-brac) domain of these proteins presents a remarkable structural versatility, being able to adopt a variety of oligomeric states. To gain insights into this intriguing aspect, we performed extensive molecular dynamics simulations on several BTB domains of KCTD proteins in different oligomeric states (monomers, dimers, tetramers, and open/close pentamers). These studies indicate that KCTD-BTB domains are stable in the simulation timescales, even in their monomeric forms. Moreover, simulations also show that the dynamic behavior of open pentameric states is strictly related to their functional roles and that different KCTDs may form stable hetero-oligomers. Molecular dynamics (MD) simulations also provided a dynamic view of the complex formed by KCTD16 and the GABAB2 receptor, whose structure has been recently reported. Finally, simulations carried out on the isolated fragment of the GABAB2 receptor that binds KCTD16 indicate that it is able to assume the local conformation required for the binding to KCTD.
Collapse
|
26
|
Smaldone G, Balasco N, Pirone L, Caruso D, Di Gaetano S, Pedone EM, Vitagliano L. Molecular basis of the scalp-ear-nipple syndrome unraveled by the characterization of disease-causing KCTD1 mutants. Sci Rep 2019; 9:10519. [PMID: 31324836 PMCID: PMC6642198 DOI: 10.1038/s41598-019-46911-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/05/2019] [Indexed: 12/11/2022] Open
Abstract
The scalp-ear-nipple (SEN) syndrome is an autosomal-dominant disorder characterized by cutis aplasia of the scalp and malformations of breast, external ears, digits, and nails. Genetic analyses have shown that the disease is caused by missense mutations of the KCTD1 protein, although the functional/structural basis of SEN insurgence is hitherto unknown. With the aim of unravelling the molecular basis of the SEN syndrome associated with KCTD1 mutations we here expressed and characterized several disease causing mutants. A preliminary dissection of the protein provides insights into the role that individual domains play in KCTD1 stability. The characterization of SEN-causing mutants indicates that, although the mutation sites are located in distant regions of the BTB domain or of the pre-BTB region, all of them are unable to interact with the transcription factor AP-2α, a well-known KCTD1 biological partner. Notably, all mutations, including the one located in the pre-BTB region, produce a significant destabilization of the protein. The structural role of the pre-BTB region in KCTD1 and other proteins of the family is corroborated by its sequence conservation in orthologs and paralogs. Interestingly, SEN-causing mutations also favor the tendency of KCTD1 to adopt structural states that are characterized by the ability to bind the β-amyloid fluorescent dye thioflavin T. The formation of aggregation-prone species may have important implications for the disease etiology. Collectively, these findings provide an intriguing picture of the functional and structural alterations induced by KCTD1 mutations that ultimately lead to disease.
Collapse
Affiliation(s)
| | - Nicole Balasco
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Daniela Caruso
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134, Napoli, Italy.,Università degli Studi della Campania "Luigi Vanvitelli", Viale Abramo Lincoln 5, 81100, Caserta, Italy
| | - Sonia Di Gaetano
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Emilia Maria Pedone
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134, Napoli, Italy.
| |
Collapse
|
27
|
Teng X, Aouacheria A, Lionnard L, Metz KA, Soane L, Kamiya A, Hardwick JM. KCTD: A new gene family involved in neurodevelopmental and neuropsychiatric disorders. CNS Neurosci Ther 2019; 25:887-902. [PMID: 31197948 PMCID: PMC6566181 DOI: 10.1111/cns.13156] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
The underlying molecular basis for neurodevelopmental or neuropsychiatric disorders is not known. In contrast, mechanistic understanding of other brain disorders including neurodegeneration has advanced considerably. Yet, these do not approach the knowledge accrued for many cancers with precision therapeutics acting on well-characterized targets. Although the identification of genes responsible for neurodevelopmental and neuropsychiatric disorders remains a major obstacle, the few causally associated genes are ripe for discovery by focusing efforts to dissect their mechanisms. Here, we make a case for delving into mechanisms of the poorly characterized human KCTD gene family. Varying levels of evidence support their roles in neurocognitive disorders (KCTD3), neurodevelopmental disease (KCTD7), bipolar disorder (KCTD12), autism and schizophrenia (KCTD13), movement disorders (KCTD17), cancer (KCTD11), and obesity (KCTD15). Collective knowledge about these genes adds enhanced value, and critical insights into potential disease mechanisms have come from unexpected sources. Translation of basic research on the KCTD-related yeast protein Whi2 has revealed roles in nutrient signaling to mTORC1 (KCTD11) and an autophagy-lysosome pathway affecting mitochondria (KCTD7). Recent biochemical and structure-based studies (KCTD12, KCTD13, KCTD16) reveal mechanisms of regulating membrane channel activities through modulation of distinct GTPases. We explore how these seemingly varied functions may be disease related.
Collapse
Affiliation(s)
- Xinchen Teng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyJohns Hopkins University Bloomberg School of Public HealthBaltimoreMaryland
| | - Abdel Aouacheria
- ISEM, Institut des Sciences de l'Evolution de Montpellier, CNRS, EPHE, IRDUniversité de MontpellierMontpellierFrance
| | - Loïc Lionnard
- ISEM, Institut des Sciences de l'Evolution de Montpellier, CNRS, EPHE, IRDUniversité de MontpellierMontpellierFrance
| | - Kyle A. Metz
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyJohns Hopkins University Bloomberg School of Public HealthBaltimoreMaryland
- Present address:
Feinberg School of MedicineNorthwestern UniversityChicagoUSA
| | - Lucian Soane
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyJohns Hopkins University Bloomberg School of Public HealthBaltimoreMaryland
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral SciencesJohns Hopkins School of MedicineBaltimoreMaryland
| | - J. Marie Hardwick
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyJohns Hopkins University Bloomberg School of Public HealthBaltimoreMaryland
| |
Collapse
|
28
|
Alevy J, Burger CA, Albrecht NE, Jiang D, Samuel MA. Progressive myoclonic epilepsy-associated gene Kctd7 regulates retinal neurovascular patterning and function. Neurochem Int 2019; 129:104486. [PMID: 31175897 DOI: 10.1016/j.neuint.2019.104486] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/28/2022]
Abstract
Neuron function relies on and instructs the development and precise organization of neurovascular units that in turn support circuit activity. However, our understanding of the molecular cues that regulate this relationship remains sparse. Using a high-throughput screening pipeline, we recently identified several new regulators of vascular patterning. Among these was the potassium channel tetramerization domain-containing protein 7 (KCTD7). Mutations in KCTD7 are associated with progressive myoclonic epilepsy, but how KCTD7 regulates neural development and function remains poorly understood. To begin to identify such mechanisms, we focus on mouse retina, a tractable part of the central nervous system that contains precisely ordered neuron subtypes supported by a trilaminar vascular network. We find that deletion of Kctd7 induces defective patterning of the adult retina vascular network, resulting in increased branching, vessel length, and lacunarity. These alterations reflect early and specific defects in vessel development, as emergence of the superficial and deep vascular layers were delayed. These defects are likely due to a role for Kctd7 in inner retina neurons. Kctd7 is absent from vessels but present in neurons in the inner retina, and its deletion resulted in a corresponding increase in the number of bipolar cells in development and increased vessel branching in adults. These alterations were accompanied by retinal function deficits. Together, these data suggest that neuronal Kctd7 drives growth and patterning of the vasculature and that neurovascular interactions may participate in the pathogenesis of KCTD7-related human diseases.
Collapse
Affiliation(s)
- Jonathan Alevy
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Courtney A Burger
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Nicholas E Albrecht
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Gao C, Pallett MA, Croll TI, Smith GL, Graham SC. Molecular basis of cullin-3 (Cul3) ubiquitin ligase subversion by vaccinia virus protein A55. J Biol Chem 2019; 294:6416-6429. [PMID: 30819806 PMCID: PMC6484134 DOI: 10.1074/jbc.ra118.006561] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/26/2019] [Indexed: 12/22/2022] Open
Abstract
BTB-Kelch proteins are substrate-specific adaptors for cullin-3 (Cul3) RING-box-based E3 ubiquitin ligases, mediating protein ubiquitylation for subsequent proteasomal degradation. Vaccinia virus encodes three BTB-Kelch proteins: A55, C2, and F3. Viruses lacking A55 or C2 have altered cytopathic effects in cultured cells and altered pathology in vivo Previous studies have shown that the ectromelia virus orthologue of A55 interacts with Cul3 in cells. We report that the N-terminal BTB-BACK (BB) domain of A55 binds directly to the Cul3 N-terminal domain (Cul3-NTD), forming a 2:2 complex in solution. We solved the structure of an A55BB/Cul3-NTD complex from anisotropic crystals diffracting to 2.3/3.7 Å resolution in the best/worst direction, revealing that the overall interaction and binding interface closely resemble the structures of cellular BTB/Cul3-NTD complexes, despite low sequence identity between A55 and cellular BTB domains. Surprisingly, despite this structural similarity, the affinity of Cul3-NTD for A55BB was stronger than for cellular BTB proteins. Glutamate substitution of the A55 residue Ile-48, adjacent to the canonical φX(D/E) Cul3-binding motif, reduced affinity of A55BB for Cul3-NTD by at least 2 orders of magnitude. Moreover, Ile-48 and the φX(D/E) motif are conserved in A55 orthologues from other poxviruses, but not in the vaccinia virus proteins C2 or F3. The high-affinity interaction between A55BB and Cul3-NTD suggests that, in addition to directing the Cul3-RING E3 ligase complex to degrade cellular/viral target proteins that are normally unaffected, A55 may also sequester Cul3 from cellular adaptor proteins, thereby protecting substrates of these cellular adaptors from ubiquitylation and degradation.
Collapse
Affiliation(s)
- Chen Gao
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP and
| | - Mitchell A Pallett
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP and
| | - Tristan I Croll
- the Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge CB2 0XY, United Kingdom
| | - Geoffrey L Smith
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP and
| | - Stephen C Graham
- From the Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP and
| |
Collapse
|
30
|
The essential player in adipogenesis GRP78 is a novel KCTD15 interactor. Int J Biol Macromol 2018; 115:469-475. [PMID: 29665387 DOI: 10.1016/j.ijbiomac.2018.04.078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/10/2018] [Accepted: 04/13/2018] [Indexed: 01/08/2023]
Abstract
KCTD15 is a member of the K+ Channel Tetramerization Domain family, implicated in crucial physio-pathological processes. Recent evidences suggest that KCTD15 is an obesity-linked protein in humans and its Drosophila homologue is involved in food uptake. KCTD15 molecular mechanism in these processes is still unknown. To fill this gap, KCTD15 was biophysically characterized showing a folded, pentameric region endowed with a remarkable thermal stability. Notably, the C-terminal domain significantly contributes to the stabilization of the BTB N-terminal domain. The availability of large amount of stable recombinant protein also made possible a functional proteomic approach in 3T3-L1 cells to search for novel KCTD15 interactors. These investigations led to the discovery that GRP78 is a KCTD15 partner in all the adipogenesis phases. Our data clearly prove the physical interaction of the two proteins and also indicate that GRP78 plays an active role in the stabilization of KCTD15. Furthermore, the presence in Drosophila of a GRP78 homologue corroborates the physiological role played by the complex KCTD15-GRP78 in the adipogenesis process and indicates that it is evolutionarily conserved. Present results also suggest that KCTD15 may be a new target for obesity control.
Collapse
|
31
|
Wong TCB, Rebbert M, Wang C, Chen X, Heffer A, Zarelli VE, Dawid IB, Zhao H. Genes regulated by potassium channel tetramerization domain containing 15 (Kctd15) in the developing neural crest. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2018; 60:159-66. [PMID: 27389986 DOI: 10.1387/ijdb.160058id] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neural crest (NC) development is controlled precisely by a regulatory network with multiple signaling pathways and the involvement of many genes. The integration and coordination of these factors are still incompletely understood. Overexpression of Wnt3a and the BMP antagonist Chordin in animal cap cells from Xenopus blastulae induces a large number of NC specific genes. We previously suggested that Potassium Channel Tetramerization Domain containing 15 (Kctd15) regulates NC formation by affecting Wnt signaling and the activity of transcription factor AP-2. In order to advance understanding of the function of Kctd15 during NC development, we performed DNA microarray assays in explants injected with Wnt3a and Chordin, and identified genes that are affected by Kctd15 overexpression. Among the many genes identified, we chose Duf domain containing protein 1 (ddcp1), Platelet-Derived Growth Factor Receptor a (pdgfra), Complement factor properdin (cfp), Zinc Finger SWIM-Type Containing 5 (zswim5), and complement component 3 (C3) to examine their expression by whole mount in situ hybridization. Our work points to a possible role for Kctd15 in the regulation of NC formation and other steps in embryonic development.
Collapse
Affiliation(s)
- Thomas C B Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Gaglione R, Smaldone G, Di Girolamo R, Piccoli R, Pedone E, Arciello A. Cell milieu significantly affects the fate of AApoAI amyloidogenic variants: predestination or serendipity? Biochim Biophys Acta Gen Subj 2017; 1862:377-384. [PMID: 29174954 DOI: 10.1016/j.bbagen.2017.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Specific apolipoprotein A-I variants are associated to severe hereditary amyloidoses. The organ distribution of AApoAI amyloidosis seems to depend on the position of the mutation, since mutations in residues from 1 to 75 are mainly associated to hepatic and renal amyloidosis, while mutations in residues from 173 to 178 are mostly responsible for cardiac, laryngeal, and cutaneous amyloidosis. Molecular bases of this tissue specificity are still poorly understood, but it is increasingly emerging that protein destabilization induced by amyloidogenic mutations is neither necessary nor sufficient for amyloidosis development. METHODS By using a multidisciplinary approach, including circular dichroism, dynamic light scattering, spectrofluorometric and atomic force microscopy analyses, the effect of target cells on the conformation and fibrillogenic pathway of the two AApoAI amyloidogenic variants AApoAIL75P and AApoAIL174S has been monitored. RESULTS Our data show that specific cell milieus selectively affect conformation, aggregation propensity and fibrillogenesis of the two AApoAI amyloidogenic variants. CONCLUSIONS An intriguing picture emerged indicating that defined cell contexts selectively induce fibrillogenesis of specific AApoAI variants. GENERAL SIGNIFICANCE An innovative methodological approach, based on the use of whole intact cells to monitor the effects of cell context on AApoAI variants fibrillogenic pathway, has been set up.
Collapse
Affiliation(s)
- Rosa Gaglione
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | | | - Rocco Di Girolamo
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Renata Piccoli
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; Istituto Nazionale di Biostrutture e Biosistemi (INBB), Italy
| | - Emilia Pedone
- Istituto di Biostrutture e Bioimmagini, CNR, Naples, Italy; Research Centre on Bioactive Peptides (CIRPeB), University of Naples Federico II, Via Mezzocannone 16, 80134 Naples, Italy.
| | - Angela Arciello
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; Istituto Nazionale di Biostrutture e Biosistemi (INBB), Italy.
| |
Collapse
|
33
|
Pinkas DM, Sanvitale CE, Bufton JC, Sorrell FJ, Solcan N, Chalk R, Doutch J, Bullock AN. Structural complexity in the KCTD family of Cullin3-dependent E3 ubiquitin ligases. Biochem J 2017; 474:3747-3761. [PMID: 28963344 PMCID: PMC5664961 DOI: 10.1042/bcj20170527] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/21/2017] [Accepted: 09/25/2017] [Indexed: 12/25/2022]
Abstract
Members of the potassium channel tetramerization domain (KCTD) family are soluble non-channel proteins that commonly function as Cullin3 (Cul3)-dependent E3 ligases. Solution studies of the N-terminal BTB domain have suggested that some KCTD family members may tetramerize similarly to the homologous tetramerization domain (T1) of the voltage-gated potassium (Kv) channels. However, available structures of KCTD1, KCTD5 and KCTD9 have demonstrated instead pentameric assemblies. To explore other phylogenetic clades within the KCTD family, we determined the crystal structures of the BTB domains of a further five human KCTD proteins revealing a rich variety of oligomerization architectures, including monomer (SHKBP1), a novel two-fold symmetric tetramer (KCTD10 and KCTD13), open pentamer (KCTD16) and closed pentamer (KCTD17). While these diverse geometries were confirmed by small-angle X-ray scattering (SAXS), only the pentameric forms were stable upon size-exclusion chromatography. With the exception of KCTD16, all proteins bound to Cul3 and were observed to reassemble in solution as 5 : 5 heterodecamers. SAXS data and structural modelling indicate that Cul3 may stabilize closed BTB pentamers by binding across their BTB-BTB interfaces. These extra interactions likely also allow KCTD proteins to bind Cul3 without the expected 3-box motif. Overall, these studies reveal the KCTD family BTB domain to be a highly versatile scaffold compatible with a range of oligomeric assemblies and geometries. This observed interface plasticity may support functional changes in regulation of this unusual E3 ligase family.
Collapse
Affiliation(s)
- Daniel M Pinkas
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Caroline E Sanvitale
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Joshua C Bufton
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Fiona J Sorrell
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Nicolae Solcan
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Rod Chalk
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - James Doutch
- ISIS Pulsed Neutron and Muon Source, STFC, Harwell Science and Innovation Campus, Didcot OX11 0QX, U.K
| | - Alex N Bullock
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| |
Collapse
|
34
|
Álvarez A, Uribe F, Canales J, Romero C, Soza A, Peña MA, Antonelli M, Almarza O, Cerda O, Toledo H. KCTD5 and Ubiquitin Proteasome Signaling Are Required for Helicobacter pylori Adherence. Front Cell Infect Microbiol 2017; 7:450. [PMID: 29114497 PMCID: PMC5660694 DOI: 10.3389/fcimb.2017.00450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022] Open
Abstract
In order to establish infection, bacterial pathogens modulate host cellular processes by using virulence factors, which are delivered from the bacteria to the host cell leading to cellular reprogramming. In this context, several pathogens regulate the ubiquitin proteasome system in order to regulate the cellular effectors required for their successful colonization and persistance. In this study, we investigated how Helicobacter pylori affect the ubiquitination of the host proteins to achieve the adherence to the cells, using AGS gastric epithelial cells cultured with H. pylori strains, H. pylori 26695 and two isogenic mutants H. pylori cag::cat and vacA::apha3, to characterize the ability of H. pylori to reprogram the ubiquitin proteasome systems. The infection assays suggest that the ubiquitination of the total proteins does not change when cells were co-culture with H. pylori. We also found that the proteasome activity is necessary for H. pylori adhesion to AGS cells and the adherence increases when the level of KCTD5, an adaptor of Cullin-3, decrease. Moreover, we found that KCTD5 is ubiquitinated and degraded by the proteasome system and that CagA and VacA played no role on reducing KCTD5 levels. Furthermore, H. pylori impaired KCTD5 ubiquitination and did not increase global proteasome function. These results suggest that H. pylori affect the ubiquitin-proteasome system (UPS) to facilitate the adhesion of this microorganism to establish stable colonization in the gastric epithelium and improve our understanding of how H. pylori hijack host systems to establish the adherence.
Collapse
Affiliation(s)
- Alhejandra Álvarez
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Felipe Uribe
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Jimena Canales
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Cristóbal Romero
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Andrea Soza
- Department of Biological and Chemical Sciences, Faculty of Science, Universidad San Sebastián, Santiago, Chile
| | - María A Peña
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Marcelo Antonelli
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Oscar Almarza
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Oscar Cerda
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile
| | - Héctor Toledo
- Molecular and Cellular Biology Program, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
35
|
Genetic wiring maps of single-cell protein states reveal an off-switch for GPCR signalling. Nature 2017; 546:307-311. [DOI: 10.1038/nature22376] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/13/2017] [Indexed: 01/05/2023]
|
36
|
Blondelle J, Shapiro P, Domenighetti AA, Lange S. Cullin E3 Ligase Activity Is Required for Myoblast Differentiation. J Mol Biol 2017; 429:1045-1066. [PMID: 28238764 DOI: 10.1016/j.jmb.2017.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 01/06/2023]
Abstract
The role of cullin E3-ubiquitin ligases for muscle homeostasis is best known during muscle atrophy, as the cullin-1 substrate adaptor atrogin-1 is among the most well-characterized muscle atrogins. We investigated whether cullin activity was also crucial during terminal myoblast differentiation and aggregation of acetylcholine receptors for the establishment of neuromuscular junctions in vitro. The activity of cullin E3-ligases is modulated through post-translational modification with the small ubiquitin-like modifier nedd8. Using either the Nae1 inhibitor MLN4924 (Pevonedistat) or siRNA against nedd8 in early or late stages of differentiation on C2C12 myoblasts, and primary satellite cells from mouse and human, we show that cullin E3-ligase activity is necessary for each step of the muscle cell differentiation program in vitro. We further investigate known transcriptional repressors for terminal muscle differentiation, namely ZBTB38, Bhlhe41, and Id1. Due to their identified roles for terminal muscle differentiation, we hypothesize that the accumulation of these potential cullin E3-ligase substrates may be partially responsible for the observed phenotype. MLN4924 is currently undergoing clinical trials in cancer patients, and our experiments highlight concerns on the homeostasis and regenerative capacity of muscles in these patients who often experience cachexia.
Collapse
Affiliation(s)
- Jordan Blondelle
- Division of Cardiology, University of California San Diego, La Jolla, CA-92093 USA
| | - Paige Shapiro
- Division of Cardiology, University of California San Diego, La Jolla, CA-92093 USA
| | - Andrea A Domenighetti
- Rehabilitation Institute of Chicago, Chicago, IL-60611 USA; Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL-60611, USA
| | - Stephan Lange
- Division of Cardiology, University of California San Diego, La Jolla, CA-92093 USA.
| |
Collapse
|
37
|
Vriend J, Liu W, Reiter RJ. The pineal gland: A model for adrenergic modulation of ubiquitin ligases. PLoS One 2017; 12:e0172441. [PMID: 28212404 PMCID: PMC5315301 DOI: 10.1371/journal.pone.0172441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 02/04/2017] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION A recent study of the pineal gland of the rat found that the expression of more than 3000 genes showed significant day/night variations (The Hartley dataset). The investigators of this report made available a supplemental table in which they tabulated the expression of many genes that they did not discuss, including those coding for components of the ubiquitin proteasome system. Herein we identify the genes of the ubiquitin proteasome system whose expression were significantly influenced by environmental lighting in the Hartley dataset, those that were stimulated by DBcAMP in pineal glands in culture, and those that were stimulated by norepinephrine. PURPOSE Using the Ubiquitin and Ubiquitin-like Conjugation Database (UUCA) we identified ubiquitin ligases and conjugases, and deubiquitinases in the Hartley dataset for the purpose of determining whether expression of genes of the ubiquitin proteasome pathway were significantly influenced by day/night variations and if these variations were regulated by autonomic innervation of the pineal gland from the superior cervical ganglia. METHODS In the Hartley experiments pineal glands groups of rats sacrificed during the day and groups sacrificed during the night were examined for gene expression. Additional groups of rats had their superior cervical ganglia removed surgically or surgically decentralized and the pineal glands likewise examined for gene expression. RESULTS The genes with at least a 2-fold day/night significant difference in expression included genes for 5 ubiquitin conjugating enzymes, genes for 58 ubiquitin E3 ligases and genes for 6 deubiquitinases. A 35-fold day/night difference was noted in the expression of the gene Sik1, which codes for a protein containing both an ubiquitin binding domain (UBD) and an ubiquitin-associated (UBA) domain. Most of the significant differences in these genes were prevented by surgical removal, or disconnection, of the superior cervical ganglia, and most were responsive, in vitro, to treatment with a cyclic AMP analog, and norepinephrine. All previously described 24-hour rhythms in the pineal require an intact sympathetic input from the superior cervical ganglia. CONCLUSIONS The Hartley dataset thus provides evidence that the pineal gland is a highly useful model for studying adrenergically dependent mechanisms regulating variations in ubiquitin ligases, ubiquitin conjugases, and deubiquitinases, mechanisms that may be physiologically relevant not only in the pineal gland, but in all adrenergically innervated tissue.
Collapse
Affiliation(s)
- Jerry Vriend
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Wenjun Liu
- Department of Pathology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas, United States of America
| |
Collapse
|
38
|
KCTD2, an adaptor of Cullin3 E3 ubiquitin ligase, suppresses gliomagenesis by destabilizing c-Myc. Cell Death Differ 2017; 24:649-659. [PMID: 28060381 DOI: 10.1038/cdd.2016.151] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/09/2016] [Accepted: 12/01/2016] [Indexed: 11/08/2022] Open
Abstract
Cullin3 E3 ubiquitin ligase ubiquitinates a wide range of substrates through substrate-specific adaptors Bric-a-brac, Tramtrack, and Broad complex (BTB) domain proteins. These E3 ubiquitin ligase complexes are involved in diverse cellular functions. Our recent study demonstrated that decreased Cullin3 expression induces glioma initiation and correlates with poor prognosis of patients with malignant glioma. However, the substrate recognition mechanism associated with tumorigenesis is not completely understood. Through yeast two-hybrid screening, we identified potassium channel tetramerization domain-containing 2 (KCTD2) as a BTB domain protein that binds to Cullin3. The interaction of Cullin3 and KCTD2 was verified using immunoprecipitation and immunofluorescence. Of interest, KCTD2 expression was markedly decreased in patient-derived glioma stem cells (GSCs) compared with non-stem glioma cells. Depletion of KCTD2 using a KCTD2-specific short-hairpin RNA in U87MG glioma cells and primary Ink4a/Arf-deficient murine astrocytes markedly increased self-renewal activity in addition with an increased expression of stem cell markers, and mouse in vivo intracranial tumor growth. As an underlying mechanism for these KCTD2-mediated phenotypic changes, we demonstrated that KCTD2 interacts with c-Myc, which is a key stem cell factor, and causes c-Myc protein degradation by ubiquitination. As a result, KCTD2 depletion acquires GSC features and affects aerobic glycolysis via expression changes in glycolysis-associated genes through c-Myc protein regulation. Of clinical significance was our finding that patients having a profile of KCTD2 mRNA-low and c-Myc gene signature-high, but not KCTD2 mRNA-low and c-Myc mRNA-high, are strongly associated with poor prognosis. This study describes a novel regulatory mode of c-Myc protein in malignant gliomas and provides a potential framework for glioma therapy by targeting c-Myc function.
Collapse
|
39
|
Heride C, Rigden DJ, Bertsoulaki E, Cucchi D, De Smaele E, Clague MJ, Urbé S. The centrosomal deubiquitylase USP21 regulates Gli1 transcriptional activity and stability. J Cell Sci 2016; 129:4001-4013. [PMID: 27621083 PMCID: PMC5117204 DOI: 10.1242/jcs.188516] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/05/2016] [Indexed: 12/23/2022] Open
Abstract
USP21 is a centrosome-associated deubiquitylase (DUB) that has been implicated in the formation of primary cilia – crucial organelles for the regulation of the Hedgehog (Hh) signaling pathway in vertebrates. Here, we identify KCTD6 – a cullin-3 E3-ligase substrate adapter that has been previously linked to Hh signaling – as well as Gli1, the key transcription factor responsible for Hh signal amplification, as new interacting partners of USP21. We identify a cryptic structured protein interaction domain in KCTD6, which is predicted to have a similar fold to Smr domains. Importantly, we show that both depletion and overexpression of catalytically active USP21 suppress Gli1-dependent transcription. Gli proteins are negatively regulated through protein kinase A (PKA)-dependent phosphorylation. We provide evidence that USP21 recruits and stabilises Gli1 at the centrosome where it promotes its phosphorylation by PKA. By revealing an intriguing functional pairing between a spatially restricted deubiquitylase and a kinase, our study highlights the centrosome as an important hub for signal coordination. Summary: We identify a Hedgehog-pathway-associated ubiquitin ligase adapter as a direct interaction partner of the deubiquitylase USP21 and discover a close interplay between USP21 and protein kinase A in regulating Gli1.
Collapse
Affiliation(s)
- Claire Heride
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Daniel J Rigden
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Erithelgi Bertsoulaki
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Danilo Cucchi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Michael J Clague
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Sylvie Urbé
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| |
Collapse
|
40
|
Smaldone G, Pirone L, Pedone E, Marlovits T, Vitagliano L, Ciccarelli L. The BTB domains of the potassium channel tetramerization domain proteins prevalently assume pentameric states. FEBS Lett 2016; 590:1663-71. [PMID: 27152988 DOI: 10.1002/1873-3468.12203] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 01/05/2023]
Abstract
Potassium channel tetramerization domain-containing (KCTD) proteins are involved in fundamental physio-pathological processes. Here, we report an analysis of the oligomeric state of the Bric-à-brack, Tram-track, Broad complex (BTB) domains of seven distinct KCTDs belonging to five major clades of the family evolution tree. Despite their functional and sequence variability, present electron microscopy data highlight the occurrence of well-defined pentameric states for all domains. Our data also show that these states coexist with alternative forms which include open pentamers. Thermal denaturation analyses conducted using KCTD1 as a model suggest that, in these proteins, different domains cooperate to their overall stability. Finally, negative-stain electron micrographs of KCTD6(BTB) in complex with Cullin3 show the presence of assemblies with a five-pointed pinwheel shape.
Collapse
Affiliation(s)
| | - Luciano Pirone
- Institute of Biostructures and Bioimaging, C.N.R., Napoli, Italy.,Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (C.I.R.C.M.S.B.), Catania, Italy
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, C.N.R., Napoli, Italy
| | - Thomas Marlovits
- Center for Structural Systems Biology (CSSB), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Deutsches Elektronen-Synchrotron (DESY), Hamburg, Germany.,Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna, Austria.,Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, C.N.R., Napoli, Italy
| | - Luciano Ciccarelli
- Center for Structural Systems Biology (CSSB), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Deutsches Elektronen-Synchrotron (DESY), Hamburg, Germany.,Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna, Austria.,Research Institute of Molecular Pathology (IMP), Vienna, Austria
| |
Collapse
|
41
|
Barone D, Balasco N, Vitagliano L. KCTD5 is endowed with large, functionally relevant, interdomain motions. J Biomol Struct Dyn 2015; 34:1725-35. [PMID: 26336981 DOI: 10.1080/07391102.2015.1090343] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The KCTD family is an emerging class of proteins that are involved in important biological processes whose biochemical and structural properties are rather poorly characterized or even completely undefined. We here used KCTD5, the only member of the family with a known three-dimensional structure, to gain insights into the intrinsic structural stability of the C-terminal domain (CTD) and into the mutual dynamic interplay between the two domains of the protein. Molecular dynamics (MD) simulations indicate that in the simulation timescale (120 ns), the pentameric assembly of the CTD is endowed with a significant intrinsic stability. Moreover, MD analyses also led to the identification of exposed β-strand residues. Being these regions intrinsically sticky, they could be involved in the substrate recognition. More importantly, simulations conducted on the full-length protein provide interesting information of the relative motions between the BTB domain and the CTD of the protein. Indeed, the dissection of the overall motion of the protein is indicative of a large interdomain twisting associated with limited bending movements. Notably, MD data indicate that the entire interdomain motion is pivoted by a single residue (Ser150) of the hinge region that connects the domains. The functional relevance of these motions was evaluated in the context of the functional macromolecular machinery in which KCTD5 is involved. This analysis indicates that the interdomain twisting motion here characterized may be important for the correct positioning of the substrate to be ubiquitinated with respect to the other factors of the ubiquitination machinery.
Collapse
Affiliation(s)
- Daniela Barone
- a Institute of Biostructures and Bioimaging, C.N.R. , Via Mezzocannone 16, Naples I-80134 , Italy.,b Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche , Seconda Università di Napoli , Caserta 81100 , Italy
| | - Nicole Balasco
- a Institute of Biostructures and Bioimaging, C.N.R. , Via Mezzocannone 16, Naples I-80134 , Italy.,b Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche , Seconda Università di Napoli , Caserta 81100 , Italy
| | - Luigi Vitagliano
- a Institute of Biostructures and Bioimaging, C.N.R. , Via Mezzocannone 16, Naples I-80134 , Italy
| |
Collapse
|
42
|
Ji AX, Chu A, Nielsen TK, Benlekbir S, Rubinstein JL, Privé GG. Structural Insights into KCTD Protein Assembly and Cullin3 Recognition. J Mol Biol 2015; 428:92-107. [PMID: 26334369 DOI: 10.1016/j.jmb.2015.08.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 02/01/2023]
Abstract
Cullin3 (Cul3)-based ubiquitin E3 ligase complexes catalyze the transfer of ubiquitin from an E2 enzyme to target substrate proteins. In these assemblies, the C-terminal region of Cul3 binds Rbx1/E2-ubiquitin, while the N-terminal region interacts with various BTB (bric-à-brac, tramtrack, broad complex) domain proteins that serve as substrate adaptors. Previous crystal structures of the homodimeric BTB proteins KLHL3, KLHL11 and SPOP in complex with the N-terminal domain of Cul3 revealed the features required for Cul3 recognition in these proteins. A second class of BTB-domain-containing proteins, the KCTD proteins, is also Cul3 substrate adaptors, but these do not share many of the previously identified determinants for Cul3 binding. We report the pentameric crystal structures of the KCTD1 and KCTD9 BTB domains and identify plasticity in the KCTD1 rings. We find that the KCTD proteins 5, 6, 9 and 17 bind to Cul3 with high affinity, while the KCTD proteins 1 and 16 do not have detectable binding. Finally, we confirm the 5:5 assembly of KCTD9/Cul3 complexes by cryo-electron microscopy and provide a molecular rationale for BTB-mediated Cul3 binding specificity in the KCTD family.
Collapse
Affiliation(s)
- Alan X Ji
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Anh Chu
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Tine Kragh Nielsen
- Princess Margaret Cancer Centre, Campbell Family Institute for Cancer Research, University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Samir Benlekbir
- The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - John L Rubinstein
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | - Gilbert G Privé
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Princess Margaret Cancer Centre, Campbell Family Institute for Cancer Research, University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON, M5G 2M9, Canada.
| |
Collapse
|