1
|
Saha A, Das S, De S, Dutta T, Roy S, Biswas A, Sengupta M. An Effort to Identify Genetic Determinants in Siblings With Wilson Disease Manifesting Striking Clinical Heterogeneity: An Exome Profiling Study of Two Indian Families. Pediatr Neurol 2024; 155:1-7. [PMID: 38552405 DOI: 10.1016/j.pediatrneurol.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Wilson disease (WD) is a rare autosomal recessive disorder of copper metabolism caused due to mutations in the copper transporter ATP7B. There is often a striking variability of clinical manifestations among patients with ATP7B mutations, including in siblings. This phenomenon may be caused by individual differences in copper accumulation in hepatocytes and intolerance to copper toxicity as governed by genetic variations in copper metabolism genes acting as modifier loci to the disease. OBJECTIVE To elucidate the genetic basis of striking clinical heterogeneity among two siblings of two families with WD. METHODS The disease diagnosis and subsequent clinical examinations were performed by expert clinicians. The younger siblings in both families presented with early neurological manifestations at a younger age than their older siblings. Interestingly, only the younger siblings were reported to have had hepatic manifestations. Exome sequencing of all the four individuals was performed to understand their heterogeneous phenotypic outcomes. RESULTS Genetic screening revealed no difference in the ATP7B variant spectrum between the siblings of each family. However, the siblings of both the families were found to harbor mutually exclusive pathogenic variants in suspected modifier genes implicated in copper metabolism and/or other neurological and hepatic disorders having overlapping symptoms with WD, viz., CFTR, PPARG, ABCB11, ATP7A, CYP2D6, mTOR, TOR1A, and CP, which can potentially explain their differential clinical phenotypes. CONCLUSION Clinical heterogeneity between siblings with WD with the same ATP7B mutation profile may be attributed to the presence of different pathogenic variants in potential modifier genes.
Collapse
Affiliation(s)
- Arpan Saha
- Department of Genetics, University of Calcutta, Kolkata, India
| | - Shristi Das
- Department of Genetics, University of Calcutta, Kolkata, India
| | - Samragni De
- Department of Genetics, University of Calcutta, Kolkata, India
| | - Tithi Dutta
- Department of Genetics, University of Calcutta, Kolkata, India
| | - Shubhrajit Roy
- The Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Atanu Biswas
- Department of Neurology, Bangur Institute of Neurosciences, Kolkata, India
| | - Mainak Sengupta
- Department of Genetics, University of Calcutta, Kolkata, India.
| |
Collapse
|
2
|
Laulumaa S, Kumpula EP, Huiskonen JT, Varjosalo M. Structure and interactions of the endogenous human Commander complex. Nat Struct Mol Biol 2024; 31:925-938. [PMID: 38459129 PMCID: PMC11189303 DOI: 10.1038/s41594-024-01246-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/19/2024] [Indexed: 03/10/2024]
Abstract
The Commander complex, a 16-protein assembly, plays multiple roles in cell homeostasis, cell cycle and immune response. It consists of copper-metabolism Murr1 domain proteins (COMMD1-10), coiled-coil domain-containing proteins (CCDC22 and CCDC93), DENND10 and the Retriever subcomplex (VPS26C, VPS29 and VPS35L), all expressed ubiquitously in the body and linked to various diseases. Here, we report the structure and key interactions of the endogenous human Commander complex by cryogenic-electron microscopy and mass spectrometry-based proteomics. The complex consists of a stable core of COMMD1-10 and an effector containing DENND10 and Retriever, scaffolded together by CCDC22 and CCDC93. We establish the composition of Commander and reveal major interaction interfaces. These findings clarify its roles in intracellular transport, and uncover a strong association with cilium assembly, and centrosome and centriole functions.
Collapse
Affiliation(s)
- Saara Laulumaa
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Esa-Pekka Kumpula
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Juha T Huiskonen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
3
|
Yao Z, Xiao Y, Li W, Kong S, Tu H, Guo S, Liu Z, Ma L, Qiao R, Wang S, Chang M, Zhao X, Zhang Y, Xu L, Sun D, Fu X. FDA-Approved Tedizolid Phosphate Prevents Cisplatin-Induced Hearing Loss Without Decreasing Its Anti-tumor Effect. J Assoc Res Otolaryngol 2024; 25:259-275. [PMID: 38622383 DOI: 10.1007/s10162-024-00945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/04/2024] [Indexed: 04/17/2024] Open
Abstract
PURPOSE Cisplatin is a low-cost clinical anti-tumor drug widely used to treat solid tumors. However, its use could damage cochlear hair cells, leading to irreversible hearing loss. Currently, there appears one drug approved in clinic only used for reducing ototoxicity associated with cisplatin in pediatric patients, which needs to further explore other candidate drugs. METHODS Here, by screening 1967 FDA-approved drugs to protect cochlear hair cell line (HEI-OC1) from cisplatin damage, we found that Tedizolid Phosphate (Ted), a drug indicated for the treatment of acute infections, had the best protective effect. Further, we evaluated the protective effect of Ted against ototoxicity in mouse cochlear explants, zebrafish, and adult mice. The mechanism of action of Ted was further explored using RNA sequencing analysis and verified. Meanwhile, we also observed the effect of Ted on the anti-tumor effect of cisplatin. RESULTS Ted had a strong protective effect on hair cell (HC) loss induced by cisplatin in zebrafish and mouse cochlear explants. In addition, when administered systemically, it protected mice from cisplatin-induced hearing loss. Moreover, antitumor studies showed that Ted had no effect on the antitumor activity of cisplatin both in vitro and in vivo. RNA sequencing analysis showed that the otoprotective effect of Ted was mainly achieved by inhibiting phosphorylation of ERK. Consistently, ERK activator aggravated the damage of cisplatin to HCs. CONCLUSION Collectively, these results showed that FDA-approved Ted protected HCs from cisplatin-induced HC loss by inhibiting ERK phosphorylation, indicating its potential as a candidate for preventing cisplatin ototoxicity in clinical settings.
Collapse
Affiliation(s)
- Zhiwei Yao
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Yu Xiao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
- School of Life Science, Shandong University, Qingdao, 266237, China
| | - Wen Li
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China.
| | - Shuhui Kong
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China
| | - Hailong Tu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Siwei Guo
- School of Life Science, Shandong University, Qingdao, 266237, China
| | - Ziyi Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Lushun Ma
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Ruifeng Qiao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China
| | - Song Wang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Miao Chang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Xiaoxu Zhao
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yuan Zhang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China.
| | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Xiaolong Fu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China.
| |
Collapse
|
4
|
You G, Zhou C, Wang L, Liu Z, Fang H, Yao X, Zhang X. COMMD proteins function and their regulating roles in tumors. Front Oncol 2023; 13:1067234. [PMID: 36776284 PMCID: PMC9910083 DOI: 10.3389/fonc.2023.1067234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
The COMMD proteins are a highly conserved protein family with ten members that play a crucial role in a variety of biological activities, including copper metabolism, endosomal sorting, ion transport, and other processes. Recent research have demonstrated that the COMMD proteins are closely associated with a wide range of disorders, such as hepatitis, myocardial ischemia, cerebral ischemia, HIV infection, and cancer. Among these, the role of COMMD proteins in tumors has been thoroughly explored; they promote or inhibit cancers such as lung cancer, liver cancer, gastric cancer, and prostate cancer. COMMD proteins can influence tumor proliferation, invasion, metastasis, and tumor angiogenesis, which are strongly related to the prognosis of tumors and are possible therapeutic targets for treating tumors. In terms of molecular mechanism, COMMD proteins in tumor cells regulate the oncogenes of NF-κB, HIF, c-MYC, and others, and are related to signaling pathways including apoptosis, autophagy, and ferroptosis. For the clinical diagnosis and therapy of malignancies, additional research into the involvement of COMMD proteins in cancer is beneficial.
Collapse
Affiliation(s)
- Guangqiang You
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China
| | - Chen Zhou
- Department of General Affairs, First Hospital of Jilin University (the Eastern Division), Jilin University, Changchun, China
| | - Lei Wang
- Department of Pediatric Neurology, First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zefeng Liu
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China
| | - He Fang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiaoxao Yao
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China,*Correspondence: Xiaoxao Yao, ; Xuewen Zhang,
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China,*Correspondence: Xiaoxao Yao, ; Xuewen Zhang,
| |
Collapse
|
5
|
Farinha CM, Gentzsch M. Revisiting CFTR Interactions: Old Partners and New Players. Int J Mol Sci 2021; 22:13196. [PMID: 34947992 PMCID: PMC8703571 DOI: 10.3390/ijms222413196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 01/07/2023] Open
Abstract
Remarkable progress in CFTR research has led to the therapeutic development of modulators that rescue the basic defect in cystic fibrosis. There is continuous interest in studying CFTR molecular disease mechanisms as not all cystic fibrosis patients have a therapeutic option available. Addressing the basis of the problem by comprehensively understanding the critical molecular associations of CFTR interactions remains key. With the availability of CFTR modulators, there is interest in comprehending which interactions are critical to rescue CFTR and which are altered by modulators or CFTR mutations. Here, the current knowledge on interactions that govern CFTR folding, processing, and stability is summarized. Furthermore, we describe protein complexes and signal pathways that modulate the CFTR function. Primary epithelial cells display a spatial control of the CFTR interactions and have become a common system for preclinical and personalized medicine studies. Strikingly, the novel roles of CFTR in development and differentiation have been recently uncovered and it has been revealed that specific CFTR gene interactions also play an important role in transcriptional regulation. For a comprehensive understanding of the molecular environment of CFTR, it is important to consider CFTR mutation-dependent interactions as well as factors affecting the CFTR interactome on the cell type, tissue-specific, and transcriptional levels.
Collapse
Affiliation(s)
- Carlos M. Farinha
- BioISI—Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016 Lisboa, Portugal
| | - Martina Gentzsch
- Marsico Lung Institute and Cystic Fibrosis Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pediatrics, Division of Pediatric Pulmonology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Laulumaa S, Varjosalo M. Commander Complex-A Multifaceted Operator in Intracellular Signaling and Cargo. Cells 2021; 10:cells10123447. [PMID: 34943955 PMCID: PMC8700231 DOI: 10.3390/cells10123447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Commander complex is a 16-protein complex that plays multiple roles in various intracellular events in endosomal cargo and in the regulation of cell homeostasis, cell cycle and immune response. It consists of COMMD1-10, CCDC22, CCDC93, DENND10, VPS26C, VPS29, and VPS35L. These proteins are expressed ubiquitously in the human body, and they have been linked to diseases including Wilson's disease, atherosclerosis, and several types of cancer. In this review we describe the function of the commander complex in endosomal cargo and summarize the individual known roles of COMMD proteins in cell signaling and cancer. It becomes evident that commander complex might be a much more important player in intracellular regulation than we currently understand, and more systematic research on the role of commander complex is required.
Collapse
|
7
|
Maung MT, Carlson A, Olea-Flores M, Elkhadragy L, Schachtschneider KM, Navarro-Tito N, Padilla-Benavides T. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J 2021; 35:e21810. [PMID: 34390520 DOI: 10.1096/fj.202100273rr] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022]
Abstract
Copper (Cu) is an essential micronutrient required for the activity of redox-active enzymes involved in critical metabolic reactions, signaling pathways, and biological functions. Transporters and chaperones control Cu ion levels and bioavailability to ensure proper subcellular and systemic Cu distribution. Intensive research has focused on understanding how mammalian cells maintain Cu homeostasis, and how molecular signals coordinate Cu acquisition and storage within organs. In humans, mutations of genes that regulate Cu homeostasis or facilitate interactions with Cu ions lead to numerous pathologic conditions. Malfunctions of the Cu+ -transporting ATPases ATP7A and ATP7B cause Menkes disease and Wilson disease, respectively. Additionally, defects in the mitochondrial and cellular distributions and homeostasis of Cu lead to severe neurodegenerative conditions, mitochondrial myopathies, and metabolic diseases. Cu has a dual nature in carcinogenesis as a promotor of tumor growth and an inducer of redox stress in cancer cells. Cu also plays role in cancer treatment as a component of drugs and a regulator of drug sensitivity and uptake. In this review, we provide an overview of the current knowledge of Cu metabolism and transport and its relation to various human pathologies.
Collapse
Affiliation(s)
- May T Maung
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Monserrat Olea-Flores
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | - Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | | |
Collapse
|
8
|
Weiskirchen R, Penning LC. COMMD1, a multi-potent intracellular protein involved in copper homeostasis, protein trafficking, inflammation, and cancer. J Trace Elem Med Biol 2021; 65:126712. [PMID: 33482423 DOI: 10.1016/j.jtemb.2021.126712] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/10/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022]
Abstract
Copper is a trace element indispensable for life, but at the same time it is implicated in reactive oxygen species formation. Several inherited copper storage diseases are described of which Wilson disease (copper overload, mutations in ATP7B gene) and Menkes disease (copper deficiency, mutations in ATP7A gene) are the most prominent ones. After the discovery in 2002 of a novel gene product (i.e. COMMD1) involved in hepatic copper handling in Bedlington terriers, studies on the mechanism of action of COMMD1 revealed numerous non-copper related functions. Effects on hepatic copper handling are likely mediated via interactions with ATP7B. In addition, COMMD1 has many more interacting partners which guide their routing to either the plasma membrane or, often in an ubiquitination-dependent fashion, trigger their proteolysis via the S26 proteasome. By stimulating NF-κB ubiquitination, COMMD1 dampens an inflammatory reaction. Finally, targeting COMMD1 function can be a novel approach in the treatment of tumors.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Louis C Penning
- Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Department of Clinical Sciences of Companion Animals, 3584 CM, Utrecht, the Netherlands.
| |
Collapse
|
9
|
Campion CG, Verissimo T, Cossette S, Tremblay J. Does Subtelomeric Position of COMMD5 Influence Cancer Progression? Front Oncol 2021; 11:642130. [PMID: 33768002 PMCID: PMC7985453 DOI: 10.3389/fonc.2021.642130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/01/2021] [Indexed: 01/04/2023] Open
Abstract
The COMMD proteins are a family of ten pleiotropic factors which are widely conserved throughout evolution and are involved in the regulation of many cellular and physiological processes. COMMD proteins are mainly expressed in adult tissue and their downregulation has been correlated with tumor progression and poor prognosis in cancer. Among this family, COMMD5 emerged as a versatile modulator of tumor progression. Its expression can range from being downregulated to highly up regulated in a variety of cancer types. Accordingly, two opposing functions could be proposed for COMMD5 in cancer. Our studies supported a role for COMMD5 in the establishment and maintenance of the epithelial cell phenotype, suggesting a tumor suppressor function. However, genetic alterations leading to amplification of COMMD5 proteins have also been observed in various types of cancer, suggesting an oncogenic function. Interestingly, COMMD5 is the only member of this family that is located at the extreme end of chromosome 8, near its telomere. Here, we review some data concerning expression and role of COMMD5 and propose a novel rationale for the potential link between the subtelomeric position of COMMD5 on chromosome 8 and its contrasting functions in cancer.
Collapse
Affiliation(s)
- Carole G Campion
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Thomas Verissimo
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Suzanne Cossette
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Johanne Tremblay
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
10
|
Corbee RJ, Penning LC. COMMD1 Exemplifies the Power of Inbred Dogs to Dissect Genetic Causes of Rare Copper-Related Disorders. Animals (Basel) 2021; 11:ani11030601. [PMID: 33668783 PMCID: PMC7996361 DOI: 10.3390/ani11030601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Wilson's Disease is a rare autosomal recessive disorder in humans, often presenting with hepatic copper overload. Finding the genetic cause of a rare disease, especially if it is related to food constituents like the trace element copper, is a Herculean task. This review describes examples of how the unique population structure of in-bred dog strains led to the discovery of a novel gene and two modifier genes involved in inherited copper toxicosis. COMMD1, after the discovery in 2002, was shown to be a highly promiscuous protein involved in copper transport, protein trafficking/degradation, regulation of virus replication, and inflammation. Mutations in the ATP7A and ATP7B proteins in Labrador retrievers and Dobermann dogs resulted in a wide variation in hepatic copper levels in these breeds. To our knowledge, numerous dog breeds with inherited copper toxicosis of unknown genetic origin exist. Therefore, the possibility that men's best friend will provide new leads in rare copper storage diseases seems realistic.
Collapse
|
11
|
Singla A, Chen Q, Suzuki K, Song J, Fedoseienko A, Wijers M, Lopez A, Billadeau DD, van de Sluis B, Burstein E. Regulation of murine copper homeostasis by members of the COMMD protein family. Dis Model Mech 2021; 14:dmm.045963. [PMID: 33262129 PMCID: PMC7803461 DOI: 10.1242/dmm.045963] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/10/2020] [Indexed: 12/28/2022] Open
Abstract
Copper is an essential transition metal for all eukaryotes. In mammals, intestinal copper absorption is mediated by the ATP7A copper transporter, whereas copper excretion occurs predominantly through the biliary route and is mediated by the paralog ATP7B. Both transporters have been shown to be recycled actively between the endosomal network and the plasma membrane by a molecular machinery known as the COMMD/CCDC22/CCDC93 or CCC complex. In fact, mutations in COMMD1 can lead to impaired biliary copper excretion and liver pathology in dogs and in mice with liver-specific Commd1 deficiency, recapitulating aspects of this phenotype. Nonetheless, the role of the CCC complex in intestinal copper absorption in vivo has not been studied, and the potential redundancy of various COMMD family members has not been tested. In this study, we examined copper homeostasis in enterocyte-specific and hepatocyte-specific COMMD gene-deficient mice. We found that, in contrast to effects in cell lines in culture, COMMD protein deficiency induced minimal changes in ATP7A in enterocytes and did not lead to altered copper levels under low- or high-copper diets, suggesting that regulation of ATP7A in enterocytes is not of physiological consequence. By contrast, deficiency of any of three COMMD genes (Commd1, Commd6 or Commd9) resulted in hepatic copper accumulation under high-copper diets. We found that each of these deficiencies caused destabilization of the entire CCC complex and suggest that this might explain their shared phenotype. Overall, we conclude that the CCC complex plays an important role in ATP7B endosomal recycling and function. Summary: Examination of copper homeostasis in enterocyte-specific and hepatocyte-specific COMMD gene-deficient mice revealed that homologs of COMMD1, which has been linked previously by genetic studies to copper regulation, also regulate copper handling in mammals.
Collapse
Affiliation(s)
- Amika Singla
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qing Chen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of General Surgery, Tongji Hospital, Tongji School of Medicine, Shanghai 200065, China
| | - Kohei Suzuki
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jie Song
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alina Fedoseienko
- Section of Molecular Genetics, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.,Division of Oncology Research, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Melinde Wijers
- Section of Molecular Genetics, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Adam Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel D Billadeau
- Division of Oncology Research, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Bart van de Sluis
- Section of Molecular Genetics, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Ezra Burstein
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
12
|
Abstract
For decades, recycling of membrane proteins has been represented in figures by arrows between the "endosome" and the plasma membrane, but recently there has been an explosion in the understanding of the mechanisms and protein complexes required to facilitate protein recycling. Here, some key discoveries will be introduced, including assigning function to a number of recently recognized protein complexes and linking their function to protein recycling. Furthermore, the importance of lipid interactions and links to diseases and epithelial polarity will be summarized.
Collapse
Affiliation(s)
- Fiona J McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Dumoulin B, Ufer C, Stehling S, Heydeck D, Kuhn H, Sofi S. Identification of the COMM-domain containing protein 1 as specific binding partner for the guanine-rich RNA sequence binding factor 1. Biochim Biophys Acta Gen Subj 2020; 1864:129678. [PMID: 32645484 DOI: 10.1016/j.bbagen.2020.129678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 06/19/2020] [Accepted: 06/27/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND The guanine-rich RNA sequence binding factor 1 (GRSF1) is an RNA-binding protein of the hnRNP H/F family, which has been implicated in erythropoiesis, regulation of the redox homeostasis, embryonic brain development, mitochondrial function and cellular senescence. The molecular basis for GRSF1-RNA interaction has extensively been studied in the past but for the time being GRSF1 binding proteins have not been identified. METHODS To search for GRSF1 binding proteins we first employed the yeast two-hybrid system and screened a cDNA library of human fetal brain for potential GRSF1 binding proteins. Subsequently, we explored the protein-protein-interaction of the recombiant proteins, carried out immunoprecipitation experiments to confirm the interaction of the native proteins in living cells and performed truncation studies to identify the protein-binding motif of GRSF1. RESULTS Using the yeast two-hybrid system we identified the COMM-domain containing protein 1 (COMMD1) as specific GRSF1 binding protein and in vitro truncation studies suggested that COMMD1 interacts with the alanine-rich domain of GRSF1. Co-immunoprecipitation strategies indicated that COMMD1-GRSF1 interaction was RNA independent and also occurred in living cells expressing the two native proteins. CONCLUSION In mammalian cells the COMM-domain containing protein 1 (COMMD1) specifically interacts with the Ala-rich domain of GRSF1 in an RNA-independent manner. GENERAL SIGNIFICANCE This is the first report describing a specific GRSF1 binding protein.
Collapse
Affiliation(s)
- Bernhard Dumoulin
- Institute of Biochemistry, Charite - University Medicine Berlin, Corporate member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany
| | - Christoph Ufer
- Institute of Biochemistry, Charite - University Medicine Berlin, Corporate member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany
| | - Sabine Stehling
- Institute of Biochemistry, Charite - University Medicine Berlin, Corporate member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany
| | - Dagmar Heydeck
- Institute of Biochemistry, Charite - University Medicine Berlin, Corporate member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany
| | - Hartmut Kuhn
- Institute of Biochemistry, Charite - University Medicine Berlin, Corporate member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany.
| | - Sajad Sofi
- University of York, Department of Biology, York YO10 5DD, United Kingdom
| |
Collapse
|
14
|
Oliva Arguelles B, Riera-Romo M, Guerra Vallespi M. Antitumour peptide based on a protein derived from the horseshoe crab: CIGB-552 a promising candidate for cancer therapy. Br J Pharmacol 2020; 177:3625-3634. [PMID: 32436254 DOI: 10.1111/bph.15132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/11/2020] [Accepted: 05/04/2020] [Indexed: 01/18/2023] Open
Abstract
Peptide-based cancer therapy has been of great interest due to the unique advantages of peptides, such as their low MW, the ability to specifically target tumour cells, easily available and low toxicity in normal tissues. Therefore, identifying and synthesizing novel peptides could provide a promising option for cancer patients. The antitumour second generation peptide, CIGB-552 has been developed as a candidate for cancer therapy. Proteomic and genomic studies have identified the intracellular protein COMMD1 as the specific target of CIGB-552. This peptide penetrates to the inside tumour cells to induce the proteasomal degradation of RelA, causing the termination of NF-κB signalling. The antitumour activity of CIGB-552 has been validated in vitro in different human cancer cell lines, as well as in vivo in syngeneic and xenograft tumour mouse models and in dogs with different types of cancers. The aim of this review is to present and discuss the experimental data obtained on the action of CIGB-552, including its mechanism of action and its therapeutic potential in human chronic diseases. This peptide is already in phase I clinical trials as antineoplastic drug and has also possible application for other inflammatory and metabolic conditions.
Collapse
Affiliation(s)
- Brizaida Oliva Arguelles
- Pharmaceutical Department, Laboratory of Tumor Biology, Centre for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mario Riera-Romo
- Pharmacology Department, Institute of Marine Sciences, Havana, Cuba
| | - Maribel Guerra Vallespi
- Pharmaceutical Department, Laboratory of Tumor Biology, Centre for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
15
|
Mallam AL, Marcotte EM. Systems-wide Studies Uncover Commander, a Multiprotein Complex Essential to Human Development. Cell Syst 2019; 4:483-494. [PMID: 28544880 DOI: 10.1016/j.cels.2017.04.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/25/2017] [Accepted: 03/23/2017] [Indexed: 11/27/2022]
Abstract
Recent mass spectrometry maps of the human interactome independently support the existence of a large multiprotein complex, dubbed "Commander." Broadly conserved across animals and ubiquitously expressed in nearly every human cell type examined thus far, Commander likely plays a fundamental cellular function, akin to other ubiquitous machines involved in expression, proteostasis, and trafficking. Experiments on individual subunits support roles in endosomal protein sorting, including the trafficking of Notch proteins, copper transporters, and lipoprotein receptors. Commander is critical for vertebrate embryogenesis, and defects in the complex and its interaction partners disrupt craniofacial, brain, and heart development. Here, we review the synergy between large-scale proteomic efforts and focused studies in the discovery of Commander, describe its composition, structure, and function, and discuss how it illustrates the power of systems biology. Based on 3D modeling and biochemical data, we draw strong parallels between Commander and the retromer cargo-recognition complex, laying a foundation for future research into Commander's role in human developmental disorders.
Collapse
Affiliation(s)
- Anna L Mallam
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Edward M Marcotte
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
16
|
Jiang Z, Chen W, Zhou J, Peng Q, Zheng H, Yuan Y, Cui H, Zhao W, Sun X, Zhou Z, Liu X. Identification of COMMD1 as a novel lamin A binding partner. Mol Med Rep 2019; 20:1790-1796. [PMID: 31257505 PMCID: PMC6625409 DOI: 10.3892/mmr.2019.10419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 05/20/2019] [Indexed: 01/28/2023] Open
Abstract
Lamin A, which is encoded by the LMNA gene, regulates gene expression and genome stability through interactions with a variety of proteins. Mutations in LMNA lead to a diverse set of inherited human diseases, collectively referred to as laminopathies. To gain insight into the protein interactions of lamin A, a yeast two-hybrid screen was conducted using the carboxy-terminus of lamin A. The screen identified copper metabolism MURR1 domain-containing 1 (COMMD1) as a novel lamin A binding partner. Colocalization experiments using fluorescent confocal microscopy revealed that COMMD1 colocalized with lamin A in 293 cells. Furthermore, the COMMD1-lamin A protein interaction was also demonstrated in co-immunoprecipitation experiments. Collectively, the present study demonstrated a physical interaction between COMMD1 and lamin A, which may aid to elucidate the mechanisms of lamin A in the aging process.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Weichun Chen
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Jing Zhou
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Qi Peng
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Huiling Zheng
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Yuan Yuan
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Hongjing Cui
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Wei Zhao
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Xuerong Sun
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Zhongjun Zhou
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Xinguang Liu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
17
|
Healy MD, Hospenthal MK, Hall RJ, Chandra M, Chilton M, Tillu V, Chen KE, Celligoi DJ, McDonald FJ, Cullen PJ, Lott JS, Collins BM, Ghai R. Structural insights into the architecture and membrane interactions of the conserved COMMD proteins. eLife 2018; 7:e35898. [PMID: 30067224 PMCID: PMC6089597 DOI: 10.7554/elife.35898] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022] Open
Abstract
The COMMD proteins are a conserved family of proteins with central roles in intracellular membrane trafficking and transcription. They form oligomeric complexes with each other and act as components of a larger assembly called the CCC complex, which is localized to endosomal compartments and mediates the transport of several transmembrane cargos. How these complexes are formed however is completely unknown. Here, we have systematically characterised the interactions between human COMMD proteins, and determined structures of COMMD proteins using X-ray crystallography and X-ray scattering to provide insights into the underlying mechanisms of homo- and heteromeric assembly. All COMMD proteins possess an α-helical N-terminal domain, and a highly conserved C-terminal domain that forms a tightly interlocked dimeric structure responsible for COMMD-COMMD interactions. The COMM domains also bind directly to components of CCC and mediate non-specific membrane association. Overall these studies show that COMMD proteins function as obligatory dimers with conserved domain architectures.
Collapse
Affiliation(s)
- Michael D Healy
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| | | | - Ryan J Hall
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| | - Mintu Chandra
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| | - Molly Chilton
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUnited Kingdom
| | - Vikas Tillu
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| | - Kai-En Chen
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| | - Dion J Celligoi
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | | | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUnited Kingdom
| | - J Shaun Lott
- School of Biological SciencesThe University of AucklandAucklandNew Zealand
| | - Brett M Collins
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| | - Rajesh Ghai
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaAustralia
| |
Collapse
|
18
|
Ware AW, Cheung TT, Rasulov S, Burstein E, McDonald FJ. Epithelial Na + Channel: Reciprocal Control by COMMD10 and Nedd4-2. Front Physiol 2018; 9:793. [PMID: 29997525 PMCID: PMC6028986 DOI: 10.3389/fphys.2018.00793] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/06/2018] [Indexed: 11/25/2022] Open
Abstract
Optimal function of the epithelial sodium channel (ENaC) in the distal nephron is key to the kidney’s long-term control of salt homeostasis and blood pressure. Multiple pathways alter ENaC cell surface populations, including correct processing and trafficking in the secretory pathway to the cell surface, and retrieval from the cell surface through ubiquitination by the ubiquitin ligase Nedd4-2, clathrin-mediated endocytosis, and sorting in the endosomal system. Members of the Copper Metabolism Murr1 Domain containing (COMMD) family of 10 proteins are known to interact with ENaC. COMMD1, 3 and 9 have been shown to down-regulate ENaC, most likely through Nedd4-2, however, the other COMMD family members remain uncharacterized. To investigate the effects of the COMMD10 protein on ENaC trafficking and function, the interaction of ENaC and COMMD10 was confirmed. Stable COMMD10 knockdown in Fischer rat thyroid epithelia decreased ENaC current and this decreased current was associated with increased Nedd4-2 protein, a known negative regulator of ENaC. However, inhibition of Nedd4-2’s ubiquitination of ENaC was only able to partially rescue the observed reduction in current. Stable COMMD10 knockdown results in defects both in endocytosis and recycling of transferrin suggesting COMMD10 likely interacts with multiple pathways to regulate ENaC and therefore could be involved in the long-term control of blood pressure.
Collapse
Affiliation(s)
- Adam W Ware
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Tanya T Cheung
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sahib Rasulov
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Ezra Burstein
- Department of Internal Medicine and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Fiona J McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Kudo E, Taura M, Suico MA, Goto H, Kai H, Okada S. Transcriptional regulation of HIV-1 host factor COMMD1 by the Sp family. Int J Mol Med 2018; 41:2366-2374. [PMID: 29336469 DOI: 10.3892/ijmm.2018.3386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 01/09/2018] [Indexed: 11/05/2022] Open
Abstract
Copper metabolism Murr1 domain containing 1 (COMMD1) has multiple functions in the regulation of protein stability at the plasma membrane and in the cytoplasm. However, the regulation of COMMD1 transcriptional has remained to be elucidated. In the present study, the 5'‑flanking region (‑1,192/+83 bp) of the human COMMD1 gene was cloned. It was observed that the COMMD1 promoter region contains GC‑rich region that has 7 putative Sp1‑binding sites via in silico analysis. The proximal promoter region at ‑289/+83 bp was required for COMMD1 basal promoter activity by deletion constructs of COMMD1 promoter. Moreover, Sp1 inhibitor, mithramycin A, suppressed basal COMMD1 promoter activity. The Sp1‑binding site (‑11/‑1 bp) in the proximal promoter region was a critical site for COMMD1 gene regulation by Sp1 and Sp3. Sp1 upregulated COMMD1 promoter activity, whereas Sp3 suppressed it. Endogenous Sp1 and Sp3 bound to the proximal promoter region of COMMD1. Taken together, Sp1 constitutively regulates the basal expression of the COMMD1 gene in human epithelial cell lines.
Collapse
Affiliation(s)
- Eriko Kudo
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto 860‑0811, Japan
| | - Manabu Taura
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto 860‑0811, Japan
| | - Mary Ann Suico
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862‑0973, Japan
| | - Hiroki Goto
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto 860‑0811, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862‑0973, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto 860‑0811, Japan
| |
Collapse
|
20
|
Li H, Pesce E, Sheppard DN, Singh AK, Pedemonte N. Therapeutic approaches to CFTR dysfunction: From discovery to drug development. J Cyst Fibros 2018; 17:S14-S21. [DOI: 10.1016/j.jcf.2017.08.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 11/29/2022]
|
21
|
Okamoto CT. Regulation of Transporters and Channels by Membrane-Trafficking Complexes in Epithelial Cells. Cold Spring Harb Perspect Biol 2017; 9:a027839. [PMID: 28246186 PMCID: PMC5666629 DOI: 10.1101/cshperspect.a027839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The vectorial secretion and absorption of fluid and solutes by epithelial cells is dependent on the polarized expression of membrane solute transporters and channels at the apical and basolateral membranes. The establishment and maintenance of this polarized expression of transporters and channels are affected by divers protein-trafficking complexes. Moreover, regulation of the magnitude of transport is often under control of physiological stimuli, again through the interaction of transporters and channels with protein-trafficking complexes. This review highlights the value in utilizing transporters and channels as cargo to characterize core trafficking machinery by which epithelial cells establish and maintain their polarized expression, and how this machinery regulates fluid and solute transport in response to physiological stimuli.
Collapse
Affiliation(s)
- Curtis T Okamoto
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089-9121
| |
Collapse
|
22
|
Riera‐Romo M. COMMD1: A Multifunctional Regulatory Protein. J Cell Biochem 2017; 119:34-51. [DOI: 10.1002/jcb.26151] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Mario Riera‐Romo
- Department of PharmacologyInstitute of Marine SciencesHavanaCuba
| |
Collapse
|
23
|
Li H, Koo Y, Mao X, Sifuentes-Dominguez L, Morris LL, Jia D, Miyata N, Faulkner RA, van Deursen JM, Vooijs M, Billadeau DD, van de Sluis B, Cleaver O, Burstein E. Endosomal sorting of Notch receptors through COMMD9-dependent pathways modulates Notch signaling. J Cell Biol 2016; 211:605-17. [PMID: 26553930 PMCID: PMC4639872 DOI: 10.1083/jcb.201505108] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
COMMD protein family member COMMD9 regulates the endosome to plasma membrane trafficking of Notch through a unique COMMD–CCDC22–CCDC93 (CCC) complex. Notch family members are transmembrane receptors that mediate essential developmental programs. Upon ligand binding, a proteolytic event releases the intracellular domain of Notch, which translocates to the nucleus to regulate gene transcription. In addition, Notch trafficking across the endolysosomal system is critical in its regulation. In this study we report that Notch recycling to the cell surface is dependent on the COMMD–CCDC22–CCDC93 (CCC) complex, a recently identified regulator of endosomal trafficking. Disruption in this system leads to intracellular accumulation of Notch2 and concomitant reduction in Notch signaling. Interestingly, among the 10 copper metabolism MURR1 domain containing (COMMD) family members that can associate with the CCC complex, only COMMD9 and its binding partner, COMMD5, have substantial effects on Notch. Furthermore, Commd9 deletion in mice leads to embryonic lethality and complex cardiovascular alterations that bear hallmarks of Notch deficiency. Altogether, these studies highlight that the CCC complex controls Notch activation by modulating its intracellular trafficking and demonstrate cargo-specific effects for members of the COMMD protein family.
Collapse
Affiliation(s)
- Haiying Li
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Yeon Koo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Xicheng Mao
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Lindsey L Morris
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Da Jia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Naoteru Miyata
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rebecca A Faulkner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905 Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Marc Vooijs
- Department of Radiotherapy (MAASTRO)/GROW-School for Developmental Biology and Oncology, Maastricht University, 6229 ER Maastricht, Netherlands
| | - Daniel D Billadeau
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Bart van de Sluis
- Molecular Genetics Section - Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, Netherlands
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ezra Burstein
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390 Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
24
|
Taura M, Kudo E, Kariya R, Goto H, Matsuda K, Hattori S, Vaeteewoottacharn K, McDonald F, Suico MA, Shuto T, Kai H, Okada S. COMMD1/Murr1 reinforces HIV-1 latent infection through IκB-α stabilization. J Virol 2015; 89:2643-2658. [PMID: 25520503 PMCID: PMC4325709 DOI: 10.1128/jvi.03105-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/09/2014] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED The transcription factor NF-κB is important for HIV-1 transcription initiation in primary HIV-1 infection and reactivation in latently HIV-1-infected cells. However, comparative analysis of the regulation and function of NF-κB in latently HIV-1-infected cells has not been done. Here we show that the expression of IκB-α, an endogenous inhibitor of NF-κB, is enhanced by latent HIV-1 infection via induction of the host-derived factor COMMD1/Murr1 in myeloid cells but not in lymphoid cells by using four sets of latently HIV-1-infected cells and the respective parental cells. IκB-α protein was stabilized by COMMD1, which attenuated NF-κB signaling during Toll-like receptor ligand and tumor necrosis factor alpha treatment and enhanced HIV-1 latency in latently HIV-1-infected cells. Activation of the phosphoinositol 3-kinase (PI3K)-JAK pathway is involved in COMMD1 induction in latently HIV-1-infected cells. Our findings indicate that COMMD1 induction is the NF-κB inhibition mechanism in latently HIV-1-infected cells that contributes to innate immune deficiency and reinforces HIV-1 latency. Thus, COMMD1 might be a double-edged sword that is beneficial in primary infection but not beneficial in latent infection when HIV-1 eradication is considered. IMPORTANCE HIV-1 latency is a major barrier to viral eradication in the era of combination antiretroviral therapy. In this study, we found that COMMD1/Murr1, previously identified as an HIV-1 restriction factor, inhibits the proteasomal degradation of IκB-α by increasing the interaction with IκB-α in latently HIV-1-infected myeloid cells. IκB-α protein was stabilized by COMMD1, which attenuated NF-κB signaling during the innate immune response and enhanced HIV-1 latency in latently HIV-1-infected cells. Activation of the PI3K-JAK pathway is involved in COMMD1 induction in latently HIV-1-infected cells. Thus, the host-derived factor COMMD1 is beneficial in suppressing primary infection but enhances latent infection, indicating that it may be a double-edged sword in HIV-1 eradication.
Collapse
Affiliation(s)
- Manabu Taura
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Eriko Kudo
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Hiroki Goto
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Kouki Matsuda
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Shinichiro Hattori
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | | | - Fiona McDonald
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Mary Ann Suico
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
25
|
Phillips-Krawczak CA, Singla A, Starokadomskyy P, Deng Z, Osborne DG, Li H, Dick CJ, Gomez TS, Koenecke M, Zhang JS, Dai H, Sifuentes-Dominguez LF, Geng LN, Kaufmann SH, Hein MY, Wallis M, McGaughran J, Gecz J, Sluis BVD, Billadeau DD, Burstein E. COMMD1 is linked to the WASH complex and regulates endosomal trafficking of the copper transporter ATP7A. Mol Biol Cell 2015; 26:91-103. [PMID: 25355947 PMCID: PMC4279232 DOI: 10.1091/mbc.e14-06-1073] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/21/2014] [Accepted: 10/24/2014] [Indexed: 11/11/2022] Open
Abstract
COMMD1 deficiency results in defective copper homeostasis, but the mechanism for this has remained elusive. Here we report that COMMD1 is directly linked to early endosomes through its interaction with a protein complex containing CCDC22, CCDC93, and C16orf62. This COMMD/CCDC22/CCDC93 (CCC) complex interacts with the multisubunit WASH complex, an evolutionarily conserved system, which is required for endosomal deposition of F-actin and cargo trafficking in conjunction with the retromer. Interactions between the WASH complex subunit FAM21, and the carboxyl-terminal ends of CCDC22 and CCDC93 are responsible for CCC complex recruitment to endosomes. We show that depletion of CCC complex components leads to lack of copper-dependent movement of the copper transporter ATP7A from endosomes, resulting in intracellular copper accumulation and modest alterations in copper homeostasis in humans with CCDC22 mutations. This work provides a mechanistic explanation for the role of COMMD1 in copper homeostasis and uncovers additional genes involved in the regulation of copper transporter recycling.
Collapse
Affiliation(s)
| | | | | | - Zhihui Deng
- Department of Immunology, Department of Pathophysiology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | | | | | | | | | | | - Jin-San Zhang
- Department of Immunology, School of Pharmaceutical Sciences and Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haiming Dai
- Department of Molecular Pharmacology and Experimental Therapeutics, and
| | | | | | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, and
| | - Marco Y Hein
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Mathew Wallis
- Genetic Health Queensland at the Royal Brisbane and Women's Hospital, Herston, Queensland 4029, Australia
| | - Julie McGaughran
- Genetic Health Queensland at the Royal Brisbane and Women's Hospital, Herston, Queensland 4029, Australia School of Medicine, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jozef Gecz
- Robinson Institute and Department of Paediatrics, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Bart van de Sluis
- Section of Molecular Genetics at the Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 Groningen, Netherlands
| | - Daniel D Billadeau
- Department of Immunology, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Ezra Burstein
- Department of Internal Medicine and Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390-9151
| |
Collapse
|
26
|
Interference with ubiquitination in CFTR modifies stability of core glycosylated and cell surface pools. Mol Cell Biol 2014; 34:2554-65. [PMID: 24777605 DOI: 10.1128/mcb.01042-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is recognized that both wild-type and mutant CFTR proteins undergo ubiquitination at multiple lysines in the proteins and in one or more subcellular locations. We hypothesized that ubiquitin is added to specific sites in wild-type CFTR to stabilize it and at other sites to signal for proteolysis. Mass spectrometric analysis of wild-type CFTR identified ubiquitinated lysines 68, 710, 716, 1041, and 1080. We demonstrate that the ubiquitinated K710, K716, and K1041 residues stabilize wild-type CFTR, protecting it from proteolysis. The polyubiquitin linkage is predominantly K63. N-tail mutants, K14R and K68R, lead to increased mature band CCFTR, which can be augmented by proteasomal (but not lysosomal) inhibition, allowing trafficking to the surface. The amount of CFTR in the K1041R mutant was drastically reduced and consisted of bands A/B, suggesting that the site in transmembrane 10 (TM10) was critical to further processing beyond the proteasome. The K1218R mutant increases total and cell surface CFTR, which is further accumulated by proteasomal and lysosomal inhibition. Thus, ubiquitination at residue 1218 may destabilize wild-type CFTR in both the endoplasmic reticulum (ER) and recycling pools. Small molecules targeting the region of residue 1218 to block ubiquitination or to preserving structure at residues 710 to 716 might be protein sparing for some forms of cystic fibrosis.
Collapse
|
27
|
El Hiani Y, Linsdell P. Conformational changes opening and closing the CFTR chloride channel: insights from cysteine scanning mutagenesis. Biochem Cell Biol 2014; 92:481-8. [PMID: 25367045 DOI: 10.1139/bcb-2014-0038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cystic fibrosis, the most common lethal genetic disease affecting young people in North America, is caused by failure of the chloride ion channel known as CFTR (cystic fibrosis transmembrane conductance regulator). CFTR belongs to the large family of ATP-binding cassette (ABC) membrane transporters. In CFTR, ATP-driven events at the nucleotide-binding domains (NBDs) open and close a gate that controls chloride permeation. However, the conformational changes concomitant with opening and closing of the CFTR gate are unknown. Diverse techniques including substituted cysteine accessibility method, disulfide cross-linking, and patch-clamp recording have been used to explore CFTR channel structure. Here, we consider the architecture of both the open and the closed CFTR channel. We review how CFTR channel structure changes between the closed and the open channel conformations and portray the relative function of both cytoplasmic and vestigial gates during the gating cycle. Understanding how the CFTR channel gates chloride permeation is central for understanding how CFTR defects lead to CF. Such knowledge opens the door for novel ways to maximize CFTR channel activity in a CF setting.
Collapse
Affiliation(s)
- Yassine El Hiani
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | |
Collapse
|
28
|
Bartuzi P, Wijshake T, Dekker DC, Fedoseienko A, Kloosterhuis NJ, Youssef SA, Li H, Shiri-Sverdlov R, Kuivenhoven JA, de Bruin A, Burstein E, Hofker MH, van de Sluis B. A cell-type-specific role for murine Commd1 in liver inflammation. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2257-65. [PMID: 25072958 DOI: 10.1016/j.bbadis.2014.06.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 12/18/2022]
Abstract
The transcription factor NF-κB plays a critical role in the inflammatory response and it has been implicated in various diseases, including non-alcoholic fatty liver disease (NAFLD). Although transient NF-κB activation may protect tissues from stress, a prolonged NF-κB activation can have a detrimental effect on tissue homeostasis and therefore accurate termination is crucial. Copper Metabolism MURR1 Domain-containing 1 (COMMD1), a protein with functions in multiple pathways, has been shown to suppress NF-κB activity. However, its action in controlling liver inflammation has not yet been investigated. To determine the cell-type-specific contribution of Commd1 to liver inflammation, we used hepatocyte and myeloid-specific Commd1-deficient mice. We also used a mouse model of NAFLD to study low-grade chronic liver inflammation: we fed the mice a high fat, high cholesterol (HFC) diet, which results in hepatic lipid accumulation accompanied by liver inflammation. Depletion of hepatocyte Commd1 resulted in elevated levels of the NF-κB transactivation subunit p65 (RelA) but, surprisingly, the level of liver inflammation was not aggravated. In contrast, deficiency of myeloid Commd1 exacerbated diet-induced liver inflammation. Unexpectedly we observed that hepatic and myeloid Commd1 deficiency in the mice both augmented hepatic lipid accumulation. The elevated levels of proinflammatory cytokines in myeloid Commd1-deficient mice might be responsible for the increased level of steatosis. This increase was not seen in hepatocyte Commd1-deficient mice, in which increased lipid accumulation appeared to be independent of inflammation. Our mouse models demonstrate a cell-type-specific role for Commd1 in suppressing liver inflammation and in the progression of NAFLD.
Collapse
Affiliation(s)
- Paulina Bartuzi
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Tobias Wijshake
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Daphne C Dekker
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Alina Fedoseienko
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Niels J Kloosterhuis
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sameh A Youssef
- Dutch Molecular Pathology Center, Department of Pathology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, De Uithof, 3584 CL Utrecht, The Netherlands
| | - Haiying Li
- University of Texas Southwestern Medical Center, Departments of Internal Medicine and Molecular Biology, Dallas, TX 75390-9151, USA
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Jan-Albert Kuivenhoven
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Alain de Bruin
- Dutch Molecular Pathology Center, Department of Pathology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, De Uithof, 3584 CL Utrecht, The Netherlands
| | - Ezra Burstein
- University of Texas Southwestern Medical Center, Departments of Internal Medicine and Molecular Biology, Dallas, TX 75390-9151, USA
| | - Marten H Hofker
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Bart van de Sluis
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Molecular Genetics Section, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
29
|
Li H, Chan L, Bartuzi P, Melton SD, Weber A, Ben-Shlomo S, Varol C, Raetz M, Mao X, Starokadomskyy P, van Sommeren S, Mokadem M, Schneider H, Weisberg R, Westra HJ, Esko T, Metspalu A, Kumar V, Faubion WA, Yarovinsky F, Hofker M, Wijmenga C, Kracht M, Franke L, Aguirre V, Weersma RK, Gluck N, van de Sluis B, Burstein E. Copper metabolism domain-containing 1 represses genes that promote inflammation and protects mice from colitis and colitis-associated cancer. Gastroenterology 2014; 147:184-195.e3. [PMID: 24727021 PMCID: PMC4086320 DOI: 10.1053/j.gastro.2014.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/21/2014] [Accepted: 04/05/2014] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Activation of the transcription factor nuclear factor-κB (NF-κB) has been associated with the development of inflammatory bowel disease (IBD). Copper metabolism MURR1 domain containing 1 (COMMD1), a regulator of various transport pathways, has been shown to limit NF-κB activation. We investigated the roles of COMMD1 in the pathogenesis of colitis in mice and IBD in human beings. METHODS We created mice with a specific disruption of Commd1 in myeloid cells (Mye-knockout [K/O] mice); we analyzed immune cell populations and functions and expression of genes regulated by NF-κB. Sepsis was induced in Mye-K/O and wild-type mice by cecal ligation and puncture or intraperitoneal injection of lipopolysaccharide (LPS), colitis was induced by administration of dextran sodium sulfate, and colitis-associated cancer was induced by administration of dextran sodium sulfate and azoxymethane. We measured levels of COMMD1 messenger RNA in colon biopsy specimens from 29 patients with IBD and 16 patients without (controls), and validated findings in an independent cohort (17 patients with IBD and 22 controls). We searched for polymorphisms in or near COMMD1 that were associated with IBD using data from the International IBD Genetics Consortium and performed quantitative trait locus analysis. RESULTS In comparing gene expression patterns between myeloid cells from Mye-K/O and wild-type mice, we found that COMMD1 represses expression of genes induced by LPS. Mye-K/O mice had more intense inflammatory responses to LPS and developed more severe sepsis and colitis, with greater mortality. More Mye-K/O mice with colitis developed colon dysplasia and tumors than wild-type mice. We observed a reduced expression of COMMD1 in colon biopsy specimens and circulating leukocytes from patients with IBD. We associated single-nucleotide variants near COMMD1 with reduced expression of the gene and linked them with increased risk for ulcerative colitis. CONCLUSIONS Expression of COMMD1 by myeloid cells has anti-inflammatory effects. Reduced expression or function of COMMD1 could be involved in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Haiying Li
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Lillienne Chan
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Paulina Bartuzi
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Pediatrics, Groningen, 9713 AV, The Netherlands
| | - Shelby D. Melton
- Dallas VA Medical Center, Department of Pathology, Dallas, Texas, 75216, U.S.A
| | - Axel Weber
- Justus Liebig University, Rudolf Buchheim Institute of Pharmacology, 35392 Giessen, Germany
| | - Shani Ben-Shlomo
- Tel Aviv Sourasky Medical Center, Gastroenterology Institute, Tel Aviv, 64239, Israel
| | - Chen Varol
- Tel Aviv Sourasky Medical Center, Gastroenterology Institute, Tel Aviv, 64239, Israel
| | - Megan Raetz
- UT Southwestern Medical Center, Department of Immunology, Dallas, Texas, 75390-9151, U.S.A
| | - Xicheng Mao
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Petro Starokadomskyy
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Suzanne van Sommeren
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Gastroenterology and Hepatology, Groningen, 9713 AV, The Netherlands
| | - Mohamad Mokadem
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Heike Schneider
- Institute of Physiological Chemistry, Hannover Medical School, 30623 Hannover, Germany
| | - Reid Weisberg
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Harm-Jan Westra
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Genetics, Groningen, 9713 AV, The Netherlands
| | - Tõnu Esko
- University of Tartu, Estonian Genome Center, Tartu, 51010, Estonia
| | - Andres Metspalu
- University of Tartu, Estonian Genome Center, Tartu, 51010, Estonia
| | - Vinod Kumar
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Genetics, Groningen, 9713 AV, The Netherlands
| | - William A. Faubion
- Mayo Clinic, Division of Gastroenterology and Hepatology, Rochester, Minnesota, 55905, U.S.A
| | - Felix Yarovinsky
- UT Southwestern Medical Center, Department of Immunology, Dallas, Texas, 75390-9151, U.S.A
| | - Marten Hofker
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Pediatrics, Groningen, 9713 AV, The Netherlands
| | - Cisca Wijmenga
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Genetics, Groningen, 9713 AV, The Netherlands
| | - Michael Kracht
- Justus Liebig University, Rudolf Buchheim Institute of Pharmacology, 35392 Giessen, Germany
| | - Lude Franke
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Genetics, Groningen, 9713 AV, The Netherlands
| | - Vincent Aguirre
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, 75390-9151, U.S.A
| | - Rinse K. Weersma
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Gastroenterology and Hepatology, Groningen, 9713 AV, The Netherlands
| | - Nathan Gluck
- Tel Aviv Sourasky Medical Center, Gastroenterology Institute, Tel Aviv, 64239, Israel
| | - Bart van de Sluis
- University of Groningen, University Medical Center Groningen, Section Molecular Genetics – Department of Pediatrics, Groningen, 9713 AV, The Netherlands
| | - Ezra Burstein
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
30
|
Vonk WIM, Kakkar V, Bartuzi P, Jaarsma D, Berger R, Hofker MH, Klomp LWJ, Wijmenga C, Kampinga HH, van de Sluis B. The Copper Metabolism MURR1 domain protein 1 (COMMD1) modulates the aggregation of misfolded protein species in a client-specific manner. PLoS One 2014; 9:e92408. [PMID: 24691167 PMCID: PMC3972230 DOI: 10.1371/journal.pone.0092408] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/21/2014] [Indexed: 01/27/2023] Open
Abstract
The Copper Metabolism MURR1 domain protein 1 (COMMD1) is a protein involved in multiple cellular pathways, including copper homeostasis, NF-κB and hypoxia signalling. Acting as a scaffold protein, COMMD1 mediates the levels, stability and proteolysis of its substrates (e.g. the copper-transporters ATP7B and ATP7A, RELA and HIF-1α). Recently, we established an interaction between the Cu/Zn superoxide dismutase 1 (SOD1) and COMMD1, resulting in a decreased maturation and activation of SOD1. Mutations in SOD1, associated with the progressive neurodegenerative disorder Amyotrophic Lateral Sclerosis (ALS), cause misfolding and aggregation of the mutant SOD1 (mSOD1) protein. Here, we identify COMMD1 as a novel regulator of misfolded protein aggregation as it enhances the formation of mSOD1 aggregates upon binding. Interestingly, COMMD1 co-localizes to the sites of mSOD1 inclusions and forms high molecular weight complexes in the presence of mSOD1. The effect of COMMD1 on protein aggregation is client-specific as, in contrast to mSOD1, COMMD1 decreases the abundance of mutant Parkin inclusions, associated with Parkinson’s disease. Aggregation of a polyglutamine-expanded Huntingtin, causative of Huntington’s disease, appears unaltered by COMMD1. Altogether, this study offers new research directions to expand our current knowledge on the mechanisms underlying aggregation disease pathologies.
Collapse
Affiliation(s)
- Willianne I. M. Vonk
- University Medical Center Utrecht, Department of Metabolic and Endocrine Diseases, and Netherlands Metabolomics Center, Utrecht, the Netherlands
- University Medical Center Utrecht, Complex Genetics Section, Utrecht, the Netherlands
| | - Vaishali Kakkar
- University of Groningen, University Medical Center Groningen, Department of Cell Biology, Groningen, the Netherlands
| | - Paulina Bartuzi
- University of Groningen, University Medical Center Groningen, Molecular Genetics, Groningen, the Netherlands
| | - Dick Jaarsma
- Erasmus Medical Center, Department of Neuroscience, Rotterdam, the Netherlands
| | - Ruud Berger
- University Medical Center Utrecht, Department of Metabolic and Endocrine Diseases, and Netherlands Metabolomics Center, Utrecht, the Netherlands
| | - Marten H. Hofker
- University of Groningen, University Medical Center Groningen, Molecular Genetics, Groningen, the Netherlands
| | - Leo W. J. Klomp
- University Medical Center Utrecht, Department of Metabolic and Endocrine Diseases, and Netherlands Metabolomics Center, Utrecht, the Netherlands
| | - Cisca Wijmenga
- University Medical Center Utrecht, Complex Genetics Section, Utrecht, the Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands
| | - Harm H. Kampinga
- University of Groningen, University Medical Center Groningen, Department of Cell Biology, Groningen, the Netherlands
| | - Bart van de Sluis
- University of Groningen, University Medical Center Groningen, Molecular Genetics, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
31
|
Deregulation of COMMD1 is associated with poor prognosis in diffuse large B-cell lymphoma. PLoS One 2014; 9:e91031. [PMID: 24625556 PMCID: PMC3953211 DOI: 10.1371/journal.pone.0091031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 02/06/2014] [Indexed: 11/19/2022] Open
Abstract
Background Despite improved survival for the patients with diffuse large B-cell lymphoma (DLBCL), the prognosis after relapse is poor. The aim was to identify molecular events that contribute to relapse and treatment resistance in DLBCL. Methods We analysed 51 prospectively collected pretreatment tumour samples from clinically high risk patients treated in a Nordic phase II study with dose-dense chemoimmunotherapy and central nervous system prophylaxis with high resolution array comparative genomic hybridization (aCGH) and gene expression microarrays. Major finding was validated at the protein level immunohistochemically in a trial specific tissue microarray series of 70, and in an independent validation series of 146 patients. Results We identified 31 genes whose expression changes were strongly associated with copy number aberrations. In addition, gains of chromosomes 2p15 and 18q12.2 were associated with unfavourable survival. The 2p15 aberration harboured COMMD1 gene, whose expression had a significant adverse prognostic impact on survival. Immunohistochemical analysis of COMMD1 expression in two series confirmed the association of COMMD1 expression with poor prognosis. Conclusion COMMD1 is a potential novel prognostic factor in DLBCLs. The results highlight the value of integrated comprehensive analysis to identify prognostic markers and genetic driver events not previously implicated in DLBCL. Trial Registration ClinicalTrials.gov NCT01502982
Collapse
|
32
|
Fedoseienko A, Bartuzi P, van de Sluis B. Functional understanding of the versatile protein copper metabolism MURR1 domain 1 (COMMD1) in copper homeostasis. Ann N Y Acad Sci 2014; 1314:6-14. [PMID: 24697840 DOI: 10.1111/nyas.12353] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Copper is an important cofactor in numerous biological processes in all living organisms. However, excessive copper can be extremely toxic, so it is vital that the copper level within a cell is tightly regulated. The damaging effect of copper is seen in several hereditary forms of copper toxicity in humans and animals. At present, Wilson's disease is the best-described and best-studied copper-storage disorder in humans; it is caused by mutations in the ATP7B gene. In dogs, a mutation in the COMMD1 gene has been found to be associated with copper toxicosis. Using a liver-specific Commd1 knockout mouse, the biological role of Commd1 in copper homeostasis has been confirmed. Yet, the exact mechanism by which COMMD1 regulates copper homeostasis is still unknown. Here, we give an overview of the current knowledge and perspectives on the molecular function of COMMD1 in copper homeostasis.
Collapse
Affiliation(s)
- Alina Fedoseienko
- University of Groningen, University Medical Center Groningen, Molecular Genetics section, Groningen, the Netherlands
| | | | | |
Collapse
|
33
|
COMMD1 modulates noxious inflammation in cystic fibrosis. Int J Biochem Cell Biol 2013; 45:2402-9. [DOI: 10.1016/j.biocel.2013.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 07/12/2013] [Accepted: 07/16/2013] [Indexed: 11/23/2022]
|
34
|
Tuning NF-κB activity: a touch of COMMD proteins. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2315-21. [PMID: 24080195 DOI: 10.1016/j.bbadis.2013.09.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/06/2013] [Accepted: 09/23/2013] [Indexed: 12/28/2022]
Abstract
NF-κB is an important regulator of immunity and inflammation, and its activation pathway has been studied extensively. The mechanisms that downregulate the activity of NF-κB have also received a lot of attention, particularly since its activity needs to be terminated to prevent chronic inflammation and subsequent tissue damage. The COMMD family has been identified as a new group of proteins involved in NF-κB termination. All ten COMMD members share the structurally conserved carboxy-terminal motif, the COMM domain, and are ubiquitously expressed. They seem to play distinct and non-redundant roles in various physiological processes, including NF-κB signaling. In this review, we describe the mechanisms and proteins involved in the termination of canonical NF-κB signaling, with a specific focus on the role of the COMMD family in the down-modulation of NF-κB.
Collapse
|
35
|
McDonald FJ. COMMD1 and ion transport proteins: what is the COMMection? Focus on “COMMD1 interacts with the COOH terminus of NKCC1 in Calu-3 airway epithelial cells to modulate NKCC1 ubiquitination”. Am J Physiol Cell Physiol 2013; 305:C129-30. [DOI: 10.1152/ajpcell.00128.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
Starokadomskyy P, Gluck N, Li H, Chen B, Wallis M, Maine GN, Mao X, Zaidi IW, Hein MY, McDonald FJ, Lenzner S, Zecha A, Ropers HH, Kuss AW, McGaughran J, Gecz J, Burstein E. CCDC22 deficiency in humans blunts activation of proinflammatory NF-κB signaling. J Clin Invest 2013; 123:2244-56. [PMID: 23563313 DOI: 10.1172/jci66466] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 02/14/2013] [Indexed: 01/11/2023] Open
Abstract
NF-κB is a master regulator of inflammation and has been implicated in the pathogenesis of immune disorders and cancer. Its regulation involves a variety of steps, including the controlled degradation of inhibitory IκB proteins. In addition, the inactivation of DNA-bound NF-κB is essential for its regulation. This step requires a factor known as copper metabolism Murr1 domain-containing 1 (COMMD1), the prototype member of a conserved gene family. While COMMD proteins have been linked to the ubiquitination pathway, little else is known about other family members. Here we demonstrate that all COMMD proteins bind to CCDC22, a factor recently implicated in X-linked intellectual disability (XLID). We showed that an XLID-associated CCDC22 mutation decreased CCDC22 protein expression and impaired its binding to COMMD proteins. Moreover, some affected individuals displayed ectodermal dysplasia, a congenital condition that can result from developmental NF-κB blockade. Indeed, patient-derived cells demonstrated impaired NF-κB activation due to decreased IκB ubiquitination and degradation. In addition, we found that COMMD8 acted in conjunction with CCDC22 to direct the degradation of IκB proteins. Taken together, our results indicate that CCDC22 participates in NF-κB activation and that its deficiency leads to decreased IκB turnover in humans, highlighting an important regulatory component of this pathway.
Collapse
Affiliation(s)
- Petro Starokadomskyy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Smith L, Litman P, Liedtke CM. COMMD1 interacts with the COOH terminus of NKCC1 in Calu-3 airway epithelial cells to modulate NKCC1 ubiquitination. Am J Physiol Cell Physiol 2013; 305:C133-46. [PMID: 23515529 DOI: 10.1152/ajpcell.00394.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mice deficient in Na-K-2Cl cotransporter (NKCC1) have been generated by targeted disruption of the gene encoding NKCC1 involving the carboxy terminus (CT-NKCC1) but not the amino terminus. We hypothesize that the resulting physiological defects are due to loss of proteins interacting with CT-NKCC1. Using a yeast two-hybrid approach, adaptor protein COMMD1 was found to bind to CT-NKCC1 (aa 1,040-1,212). Binding was verified in a yeast-independent system using GST-COMMD1 and myc-CT-NKCC1. Truncated COMMD1 and CT-NKCC1 peptides were used in binding assays to identify the site of interaction. The results demonstrate concentration-dependent binding of COMMD1 (aa 1-47) to CT-NKCC1 (aa 1,040-1,134). Endogenous COMMD1 was detected in pull downs using recombinant FLAG-CT-NKCC1; this co-pull down was blocked by COMMD1 (aa 1-47). CT-NKCC1 (aa 1,040-1,137) decreased basolateral membrane expression of NKCC1, and COMMD1 (aa 1-47) increased NKCC1 membrane expression. Downregulation of COMMD1 using silencing (si)RNA led to a transient loss of endogenous COMMD1 but did not affect activation of NKCC1 by hyperosmotic sucrose. Hyperosmolarity caused a transient increase in NKCC1 membrane expression, indicating regulated trafficking of NKCC1; downregulation of COMMD1 using siRNA reduced baseline (unstimulated) NKCC1 expression and blunted a transient elevation in NKCC1 membrane expression caused by hyperosmolarity. Constitutive downregulation of COMMD1 in HT29 engineered cells exhibited loss of COMMD1 and decreased NKCC1 membrane expression with no effect on activation of NKCC1. Loss of COMMD1 in Calu-3 cells and in HT29 cells led to reduced ubiquitinated NKCC1. The results indicate a role for COMMD1 in the regulation of NKCC1 membrane expression and ubiquitination.
Collapse
Affiliation(s)
- Laura Smith
- Willard Alan Bernbaum, Center for Cystic Fibrosis Research, Departments of Pediatrics at Rainbow Babies and Children Hospital and Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | |
Collapse
|
38
|
Drévillon L, Megarbane A, Demeer B, Matar C, Benit P, Briand-Suleau A, Bodereau V, Ghoumid J, Nasser M, Decrouy X, Doco-Fenzy M, Rustin P, Gaillard D, Goossens M, Giurgea I. KBP-cytoskeleton interactions underlie developmental anomalies in Goldberg-Shprintzen syndrome. Hum Mol Genet 2013; 22:2387-99. [PMID: 23427148 DOI: 10.1093/hmg/ddt083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Goldberg-Shprintzen syndrome (GOSHS, MIM #609460) is an autosomal recessive disorder of intellectual disability, specific facial gestalt and Hirschsprung's disease (HSCR). In 2005, homozygosity mapping in a large consanguineous family identified KIAA1279 as the disease-causing gene. KIAA1279 encodes KIF-binding protein (KBP), whose function is incompletely understood. Studies have identified either the mitochondria or the cytoskeleton as the site of KBP localization and interactions. To better delineate the KIAA1279-related clinical spectrum and the molecular mechanisms involved in GOSHS, we studied five new patients from three different families. The homozygous KIAA1279 mutations in these patients (p.Arg90X, p.Ser200X or p.Arg202IlefsX2) led to nonsense-mediated mRNA decay and loss of KBP function. Despite the absence of functional KBP, respiratory chain complex activity in patient fibroblasts was normal. KBP did not co-localize with mitochondria in control human fibroblasts, but interacted with the actin and tubulin cytoskeleton. KBP expression directly affected neurite growth in a neuron-like cell line (human neuroblastoma SH-SY5Y), in keeping with the central (polymicrogyria) and enteric (HSCR) neuronal developmental defects seen in GOSHS patients. The KBP interactions with actin filaments and microtubules (MTs) demonstrated in our study constitute the first evidence that an actin MT cross-link protein is involved in neuronal development in humans.
Collapse
Affiliation(s)
- Loïc Drévillon
- Service de Biochimie et Génétique, AP-HP, Hôpital H. Mondor - A. Chenevier, Créteil F-94000, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang G, Duan DD. Regulation of activation and processing of the cystic fibrosis transmembrane conductance regulator (CFTR) by a complex electrostatic interaction between the regulatory domain and cytoplasmic loop 3. J Biol Chem 2012; 287:40484-92. [PMID: 23060444 DOI: 10.1074/jbc.m112.360214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND NEG2 regulates CFTR gating but the mechanism is unknown. RESULTS A putative NEG2-CL3 electrostatic attraction, possibly weakened by Arg-764/Arg-766 of the R domain, prohibited CFTR activation. A charge exchange between NEG2 and CL3 caused misprocessing. CONCLUSION Electrostatic regulation of CFTR activation and processing may be asymmetric at the CL3-R interface. SIGNIFICANCE The CL3-R interface is optimally designed for multiple regulations of CFTR functions. NEG2, a short C-terminal segment (817-838) of the unique regulatory (R) domain of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel, has been reported to regulate CFTR gating in response to cAMP-dependent R domain phosphorylation. The underlying mechanism, however, is unclear. Here, Lys-946 of cytoplasmic loop 3 (CL3) is proposed as counter-ion of Asp-835, Asp-836, or Glu-838 of NEG2 to prevent the channel activation by PKA. Arg-764 or Arg-766 of the Ser-768 phosphorylation site of the R domain is proposed to promote the channel activation possibly by weakening the putative CL3-NEG2 electrostatic attraction. First, not only D835A, D836A, and E838A but also K946A reduced the PKA-dependent CFTR activation. Second, both K946D and D835R/D836R/E838R mutants were activated by ATP and curcumin to a different extent. Third, R764A and R766A mutants enhanced the PKA-dependent activation. However, it is very exciting that D835R/D836R/E838R and K946D/H950D and H950R exhibited normal channel processing and activity whereas D835R/D836R/E838R/K946D/H950D was fractionally misprocessed and silent in response to forskolin. Further, D836R and E838R played a critical role in the asymmetric electrostatic regulation of CFTR processing, and Ser-768 phosphorylation may not be involved. Thus, a complex interfacial interaction among CL3, NEG2, and the Ser-768 phosphorylation site may be responsible for the asymmetric electrostatic regulation of CFTR activation and processing.
Collapse
Affiliation(s)
- Guangyu Wang
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557, USA.
| | | |
Collapse
|
40
|
El Hiani Y, Linsdell P. Role of the juxtamembrane region of cytoplasmic loop 3 in the gating and conductance of the cystic fibrosis transmembrane conductance regulator chloride channel. Biochemistry 2012; 51:3971-81. [PMID: 22545782 PMCID: PMC3381012 DOI: 10.1021/bi300065z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Opening and closing of the cystic fibrosis transmembrane conductance regulator chloride channel are controlled by interactions of ATP with its cytoplasmic nucleotide binding domains (NBDs). The NBDs are connected to the transmembrane pore via four cytoplasmic loops. These loops have been suggested to play roles both in channel gating and in forming a cytoplasmic extension of the channel pore. To investigate the structure and function of one of these cytoplasmic loops, we have used patch clamp recording to investigate the accessibility of cytoplasmically applied cysteine-reactive reagents to cysteines introduced into loop 3. We find that methanethiosulfonate (MTS) reagents modify cysteines introduced at 14 of 16 sites studied in the juxtamembrane region of loop 3, in all cases leading to inhibition of channel function. In most cases, both the functional effects of modification and the rate of modification were similar for negatively and positively charged MTS reagents. Single-channel recordings indicated that, at all sites, inhibition was the result of an MTS reagent-induced decrease in channel open probability; in no case was the Cl(-) conductance of open channels altered by modification. These results indicate that loop 3 is readily accessible to the cytoplasm and support the involvement of this region in the control of channel gating. However, our results do not support the hypothesis that this region is close enough to the Cl(-) permeation pathway to exert any influence on permeating Cl(-) ions. We propose that either the cytoplasmic pore is very wide or cytoplasmic Cl(-) ions use other routes to access the transmembrane pore.
Collapse
Affiliation(s)
- Yassine El Hiani
- Department of Physiology and Biophysics, Dalhousie University , Halifax, Nova Scotia B3H 4R2, Canada
| | | |
Collapse
|
41
|
Fieten H, Leegwater PAJ, Watson AL, Rothuizen J. Canine models of copper toxicosis for understanding mammalian copper metabolism. Mamm Genome 2012; 23:62-75. [PMID: 22147205 PMCID: PMC3275736 DOI: 10.1007/s00335-011-9378-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 11/11/2011] [Indexed: 01/13/2023]
Abstract
Hereditary forms of copper toxicosis exist in man and dogs. In man, Wilson's disease is the best studied disorder of copper overload, resulting from mutations in the gene coding for the copper transporter ATP7B. Forms of copper toxicosis for which no causal gene is known yet are recognized as well, often in young children. Although advances have been made in unraveling the genetic background of disorders of copper metabolism in man, many questions regarding disease mechanisms and copper homeostasis remain unanswered. Genetic studies in the Bedlington terrier, a dog breed affected with copper toxicosis, identified COMMD1, a gene that was previously unknown to be involved in copper metabolism. Besides the Bedlington terrier, a number of other dog breeds suffer from hereditary copper toxicosis and show similar phenotypes to humans with copper storage disorders. Unlike the heterogeneity of most human populations, the genetic structure within a purebred dog population is homogeneous, which is advantageous for unraveling the molecular genetics of complex diseases. This article reviews the work that has been done on the Bedlington terrier, summarizes what was learned from studies into COMMD1 function, describes hereditary copper toxicosis phenotypes in other dog breeds, and discusses the opportunities for genome-wide association studies on copper toxicosis in the dog to contribute to the understanding of mammalian copper metabolism and copper metabolism disorders in man.
Collapse
Affiliation(s)
- Hille Fieten
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Lukacs GL, Verkman AS. CFTR: folding, misfolding and correcting the ΔF508 conformational defect. Trends Mol Med 2011; 18:81-91. [PMID: 22138491 DOI: 10.1016/j.molmed.2011.10.003] [Citation(s) in RCA: 288] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 10/19/2011] [Accepted: 10/19/2011] [Indexed: 12/30/2022]
Abstract
Cystic fibrosis (CF), the most common lethal genetic disease in the Caucasian population, is caused by loss-of-function mutations of the CF transmembrane conductance regulator (CFTR), a cyclic AMP-regulated plasma membrane chloride channel. The most common mutation, deletion of phenylalanine 508 (ΔF508), impairs CFTR folding and, consequently, its biosynthetic and endocytic processing as well as chloride channel function. Pharmacological treatments may target the ΔF508 CFTR structural defect directly by binding to the mutant protein and/or indirectly by altering cellular protein homeostasis (proteostasis) to promote ΔF508 CFTR plasma membrane targeting and stability. This review discusses recent basic research aimed at elucidating the structural and trafficking defects of ΔF508 CFTR, a prerequisite for the rational design of CF therapy to correct the loss-of-function phenotype.
Collapse
Affiliation(s)
- Gergely L Lukacs
- Department of Physiology and GRASP, McGill University, Montréal, Quebec H3E 1Y6, Canada.
| | | |
Collapse
|
43
|
Materia S, Cater MA, Klomp LWJ, Mercer JFB, La Fontaine S. Clusterin and COMMD1 independently regulate degradation of the mammalian copper ATPases ATP7A and ATP7B. J Biol Chem 2011; 287:2485-99. [PMID: 22130675 DOI: 10.1074/jbc.m111.302216] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
ATP7A and ATP7B are copper-transporting P(1B)-type ATPases (Cu-ATPases) that are critical for regulating intracellular copper homeostasis. Mutations in the genes encoding ATP7A and ATP7B lead to copper deficiency and copper toxicity disorders, Menkes and Wilson diseases, respectively. Clusterin and COMMD1 were previously identified as interacting partners of these Cu-ATPases. In this study, we confirmed that clusterin and COMMD1 interact to down-regulate both ATP7A and ATP7B. Overexpression and knockdown of clusterin/COMMD1 decreased and increased, respectively, endogenous levels of ATP7A and ATP7B, consistent with a role in facilitating Cu-ATPase degradation. We demonstrate that whereas the clusterin/ATP7B interaction was enhanced by oxidative stress or mutation of ATP7B, the COMMD1/ATP7B interaction did not change under oxidative stress conditions, and only increased with ATP7B mutations that led to its misfolding. Clusterin and COMMD1 facilitated the degradation of ATP7B containing the same Wilson disease-causing C-terminal mutations via different degradation pathways, clusterin via the lysosomal pathway and COMMD1 via the proteasomal pathway. Furthermore, endogenous ATP7B existed in a complex with clusterin and COMMD1, but these interactions were neither competitive nor cooperative and occurred independently of each other. Together these data indicate that clusterin and COMMD1 represent alternative and independent systems regulating Cu-ATPase quality control, and consequently contributing to the maintenance of copper homeostasis.
Collapse
Affiliation(s)
- Stephanie Materia
- Strategic Research Centre for Molecular and Medical Research, School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia
| | | | | | | | | |
Collapse
|
44
|
Mao X, Gluck N, Chen B, Starokadomskyy P, Li H, Maine GN, Burstein E. COMMD1 (copper metabolism MURR1 domain-containing protein 1) regulates Cullin RING ligases by preventing CAND1 (Cullin-associated Nedd8-dissociated protein 1) binding. J Biol Chem 2011; 286:32355-65. [PMID: 21778237 PMCID: PMC3173175 DOI: 10.1074/jbc.m111.278408] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Indexed: 11/06/2022] Open
Abstract
Cullin RING ligases (CRLs), the most prolific class of ubiquitin ligase enzymes, are multimeric complexes that regulate a wide range of cellular processes. CRL activity is regulated by CAND1 (Cullin-associated Nedd8-dissociated protein 1), an inhibitor that promotes the dissociation of substrate receptor components from the CRL. We demonstrate here that COMMD1 (copper metabolism MURR1 domain-containing 1), a factor previously found to promote ubiquitination of various substrates, regulates CRL activation by antagonizing CAND1 binding. We show that COMMD1 interacts with multiple Cullins, that the COMMD1-Cul2 complex cannot bind CAND1, and that, conversely, COMMD1 can actively displace CAND1 from CRLs. These findings highlight a novel mechanism of CRL activation and suggest that CRL regulation may underlie the pleiotropic activities of COMMD1.
Collapse
Affiliation(s)
- Xicheng Mao
- From the Departments of Internal Medicine and
| | - Nathan Gluck
- the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
- the Department of Biochemistry, School of Medicine, Hebrew University, 91120 Jerusalem, Israel, and
| | - Baozhi Chen
- From the Departments of Internal Medicine and
| | | | - Haiying Li
- From the Departments of Internal Medicine and
| | - Gabriel N. Maine
- From the Departments of Internal Medicine and
- the Department of Clinical Pathology, William Beaumont Hospital, Royal Oak, Michigan 48073
| | - Ezra Burstein
- From the Departments of Internal Medicine and
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
- the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
45
|
Chang T, Ke Y, Ly K, McDonald FJ. COMMD1 regulates the delta epithelial sodium channel (δENaC) through trafficking and ubiquitination. Biochem Biophys Res Commun 2011; 411:506-11. [DOI: 10.1016/j.bbrc.2011.06.149] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 06/22/2011] [Indexed: 11/16/2022]
|