1
|
Ni Y, Liu B, Zhang W, Pang Y, Tian Y, Lv Q, Shi S, Zheng Y, Fan H. Evaluation of PDZD11 in hepatocellular carcinoma: prognostic value and diagnostic potential in combination with AFP. Front Oncol 2025; 15:1533865. [PMID: 40201341 PMCID: PMC11975663 DOI: 10.3389/fonc.2025.1533865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most prevalent liver cancer, with a 5-year survival rate below 20% and an average survival time of 3-6 months. Identifying new biomarkers is crucial for early diagnosis and prognosis. The function of PDZ domain protein 11 (PDZD11) in HCC remains unclear. Methods In this study, PDZD11 was investigated as a potential biomarker for HCC using bioinformatic analysis of the TCGA and ICGC datasets. Furthermore, we assessed the potential of serum PDZD11 as a clinical diagnostic marker by enrolling a cohort comprising 78 HCC patients and 62 healthy controls (HC) using the ELISA analysis and combining its expression with common tumor markers. Results Our research found significantly higher PDZD11 mRNA expression in HCC tissues compared to tumor-adjacent tissues (p < 0.001), which was associated with lower overall survival (OS) rates (p < 0.01). Multivariate evaluation methods established PDZD11 as a standalone predictor of prognosis. A nomogram incorporating PDZD11 expression and clinicopathological factors predicted OS rates for HCC patients over various years. Patients with HCC exhibited notably elevated serum PDZD11 levels compared to HC, with these levels rising further in advanced disease stages and deteriorating performance status (PS). ROC analysis showed high diagnostic accuracy when PDZD11 is combined with AFP (AUC = 0.958). Conclusion PDZD11 is more sensitive than AFP in assessing HCC prognosis. In conclusion, PDZD11 is a promising supplementary biomarker for HCC diagnosis and prognosis alongside AFP.
Collapse
Affiliation(s)
- Yiyun Ni
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Bin Liu
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Weizhen Zhang
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Yilin Pang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yaling Tian
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Qingsong Lv
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Shengwen Shi
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Yang Zheng
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Huihui Fan
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| |
Collapse
|
2
|
Sakurai-Yageta M, Suzuki Y. Molecular Mechanisms of Biotin in Modulating Inflammatory Diseases. Nutrients 2024; 16:2444. [PMID: 39125325 PMCID: PMC11314543 DOI: 10.3390/nu16152444] [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: 06/21/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Biotin, also known as vitamin B7 or vitamin H, is a water-soluble B-complex vitamin and serves as an essential co-enzyme for five specific carboxylases. Holocarboxylase synthase (HCS) activates biotin and facilitates its covalent attachment to these enzymes, while biotinidase releases free biotin in the biotin cycle. The transport of biotin, primarily from the intestine, is mediated by the sodium-dependent multi-vitamin transporter (SMVT). Severe biotin deficiency leads to multiple carboxylase deficiency. Moreover, biotin is crucial to glucose and lipid utilization in cellular energy production because it modulates the expression of metabolic enzymes via various signaling pathways and transcription factors. Biotin also modulates the production of proinflammatory cytokines in the immune system through similar molecular mechanisms. These regulatory roles in metabolic and immune homeostasis connect biotin to conditions such as diabetes, dermatologic manifestations, and multiple sclerosis. Furthermore, deficiencies in biotin and SMVT are implicated in inflammatory bowel disease, affecting intestinal inflammation, permeability, and flora. Notably, HCS and probably biotin directly influence gene expression through histone modification. In this review, we summarize the current knowledge on the molecular aspects of biotin and associated molecules in diseases related to both acute inflammatory responses and chronic inflammation, and discuss the potential therapeutic applications of biotin.
Collapse
Affiliation(s)
- Mika Sakurai-Yageta
- Department of Education and Training, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Miyagi, Japan
| | - Yoichi Suzuki
- Department of Education and Training, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Department of Clinical Genetics, Ageo Central General Hospital, Ageo 362-8588, Saitama, Japan
| |
Collapse
|
3
|
Chen X, Li Z, Feng Y, Yang Z, Zhao B. Identification of PDZD11 as a Potential Biomarker Associated with Immune Infiltration for Diagnosis and Prognosis in Epithelial Ovarian Cancer. Int J Gen Med 2024; 17:2113-2128. [PMID: 38766598 PMCID: PMC11102278 DOI: 10.2147/ijgm.s459418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose Evidence has indicated that PDZD11 is involved in regulating adherens junction. However, the distinct effect of its aberrant expression on epithelial ovarian cancer (EOC) awaits clarification. Methods In this study, public databases (Gene Expression Omnibus, The Cancer Genome Atlas, and The Genotype-Tissue Expression), online analysis tools (Kaplan-Meier plotter and TIMER), and data analysis methods (Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and the CIBERSORT algorithm) were fully utilized to analyze the differential expression, diagnostic efficiency, prognostic significance, potential function, and correlation with immune infiltration of PDZD11. The differential expression of PDZD11 was tested by immunohistochemistry in EOC tissues (78 cases) and control tissues (37 cases). Results Our results indicate that PDZD11 was remarkably overexpressed in EOC, which was associated with advanced cancer stages, no lymphatic metastasis status, and poor prognosis. Moreover, PDZD11 played a role in cell adhesion, cell proliferation, and immune responses. Also, PDZD11 was significantly related to the abundances of infiltrating immune cells in EOC, including neutrophils, macrophages, dendritic cells, CD8+ T cells, and CD4+ T cells, and its expression was positively co-expressed with well-known immune checkpoints, including TIGIT, TIM3, LAG3, CTLA4, and PD-1. Conclusion These results suggest that PDZD11 could be a potential diagnostic and prognostic biomarker associated with immune infiltration in EOC, and our findings might help elucidate the function of PDZD11 in carcinogenesis.
Collapse
Affiliation(s)
- Xiaoqi Chen
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhuang Li
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yanying Feng
- Department of Cardiopulmonary Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhijun Yang
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Bingbing Zhao
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
4
|
Montomoli M, Vetro A, Tubili F, Donati MA, Daniotti M, Pochiero F, Rivieri F, Girlando S, Guerrini R. A novel SLC5A6 homozygous variant in a family with multivitamin-dependent neurometabolic disorder: Phenotype expansion and long-term follow-up. Eur J Med Genet 2023:104808. [PMID: 37391029 DOI: 10.1016/j.ejmg.2023.104808] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/09/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
The sodium-dependent multivitamin transporter (hSMVT) encoded by the SLC5A6 gene is required for the intestinal absorption of biotin, pantothenic acid and lipoate, three micronutrients essential for normal growth and development. Systemic deficiency of these elements, either occurring from nutritional causes or genetic defects, is associated with neurological disorders, growth delay, skin and hair changes, metabolic and immunological abnormalities. A few patients with biallelic variants of SLC5A6 have been reported, exhibiting a spectrum of neurological and systemic clinical features with variable severity. We describe three patients from a single family carrying a homozygous p.(Leu566Valfs*33) variant of SLC5A6 disrupting the frame of the C-terminal portion of the hSMVT. In these patients, we documented a severe disorder featuring developmental delay, sensory polyneuropathy, optic atrophy, recurrent infections, and repeated episodes of intestinal pseudo-obstruction. Two patients who did not receive multivitamin supplementation therapy died in early infancy. In a third patient, early supplementation of biotin and pantothenic acid stabilized the clinical picture changing the course of the disease. These findings extend genotype-phenotype correlations and show how a timely and lifelong multivitamin treatment may be crucial to reduce the risk of life-threatening events in patients with pathogenic variants of the SLC5A6 gene.
Collapse
Affiliation(s)
- Martino Montomoli
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Annalisa Vetro
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Flavia Tubili
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Maria Alice Donati
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Marta Daniotti
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Francesca Pochiero
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | | | | | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy.
| |
Collapse
|
5
|
Rashid MA, Lin-Moshier Y, Gunaratne GS, Subramanian S, Marchant JS, Subramanian VS. Vitamin C transport in neurons and epithelia is regulated by secretory carrier-associated membrane protein-2 (SCAMP2). Int J Biol Macromol 2023; 230:123205. [PMID: 36632962 DOI: 10.1016/j.ijbiomac.2023.123205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
The human sodium-dependent vitamin C transporter-1 (hSVCT1) is localized at the apical membrane domain of polarized intestinal and renal epithelial cells to mediate ascorbic acid (AA) uptake. Currently, little is known about the array of interacting proteins that aid hSVCT1 trafficking and functional expression at the cell surface. Here we used an affinity tagging ('One-STrEP') and proteomic approach to identify hSVCT1 interacting proteins, which resolved secretory carrier-associated membrane protein-2 (SCAMP2) as a novel accessary protein partner. SCAMP2 was validated as an accessory protein by co-immunoprecipitation with hSVCT1. Co-expression of hSVCT1 and SCAMP2 in HEK-293 cells revealed both proteins co-localized in intracellular structures and at the plasma membrane. Functionally, over-expression of SCAMP2 potentiated 14C-AA uptake, and reciprocally silencing endogenous SCAMP2 decreased 14C-AA uptake. Finally, knockdown of endogenous hSVCT1 or SCAMP2 impaired differentiation of human-induced pluripotent stem cells (hiPSCs) toward a neuronal fate. These results establish SCAMP2 as a novel hSVCT1 accessary protein partner that regulates AA uptake in absorptive epithelia and during neurogenesis.
Collapse
Affiliation(s)
- Mohammad A Rashid
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, WI 53226, United States
| | - Yaping Lin-Moshier
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, WI 53226, United States
| | - Gihan S Gunaratne
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, WI 53226, United States
| | - Sreya Subramanian
- Department of Medicine, University of California, Irvine, CA 92697, United States
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, WI 53226, United States
| | | |
Collapse
|
6
|
Holling T, Nampoothiri S, Tarhan B, Schneeberger PE, Vinayan KP, Yesodharan D, Roy AG, Radhakrishnan P, Alawi M, Rhodes L, Girisha KM, Kang PB, Kutsche K. Novel biallelic variants expand the SLC5A6-related phenotypic spectrum. Eur J Hum Genet 2022; 30:439-449. [PMID: 35013551 PMCID: PMC8747999 DOI: 10.1038/s41431-021-01033-2] [Citation(s) in RCA: 11] [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: 05/14/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022] Open
Abstract
The sodium (Na+):multivitamin transporter (SMVT), encoded by SLC5A6, belongs to the sodium:solute symporter family and is required for the Na+-dependent uptake of biotin (vitamin B7), pantothenic acid (vitamin B5), the vitamin-like substance α-lipoic acid, and iodide. Compound heterozygous SLC5A6 variants have been reported in individuals with variable multisystemic disorder, including failure to thrive, developmental delay, seizures, cerebral palsy, brain atrophy, gastrointestinal problems, immunodeficiency, and/or osteopenia. We expand the phenotypic spectrum associated with biallelic SLC5A6 variants affecting function by reporting five individuals from three families with motor neuropathies. We identified the homozygous variant c.1285 A > G [p.(Ser429Gly)] in three affected siblings and a simplex patient and the maternally inherited c.280 C > T [p.(Arg94*)] variant and the paternally inherited c.485 A > G [p.(Tyr162Cys)] variant in the simplex patient of the third family. Both missense variants were predicted to affect function by in silico tools. 3D homology modeling of the human SMVT revealed 13 transmembrane helices (TMs) and Tyr162 and Ser429 to be located at the cytoplasmic facing region of TM4 and within TM11, respectively. The SLC5A6 missense variants p.(Tyr162Cys) and p.(Ser429Gly) did not affect plasma membrane localization of the ectopically expressed multivitamin transporter suggesting reduced but not abolished function, such as lower catalytic activity. Targeted therapeutic intervention yielded clinical improvement in four of the five patients. Early molecular diagnosis by exome sequencing is essential for timely replacement therapy in affected individuals.
Collapse
Affiliation(s)
- Tess Holling
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Cochin, 682041, Kerala, India
| | - Bedirhan Tarhan
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Pauline E Schneeberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Amedes MVZ Wagnerstibbe für Laboratoriumsmedizin, Hämostaseologie, Humangenetik und Mikrobiologie Hannover, 30159, Hannover, Germany
| | | | - Dhanya Yesodharan
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Cochin, 682041, Kerala, India
| | - Arun Grace Roy
- Department of Neurology, Amrita Institute of Medical Sciences & Research Centre, Cochin, 682041, Kerala, India
| | - Periyasamy Radhakrishnan
- Suma Genomics Pvt. Ltd, Manipal Universal Technology Business Incubator (MUTBI), Manipal, 576104, India
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | | | - Katta Mohan Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, 576104, India
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
- Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
7
|
Subramanian VS, Teafatiller T, Vidal J, Gunaratne GS, Rodriguez-Ortiz CJ, Kitazawa M, Marchant JS. Calsyntenin-3 interacts with the sodium-dependent vitamin C transporter-2 to regulate vitamin C uptake. Int J Biol Macromol 2021; 192:1178-1184. [PMID: 34673103 PMCID: PMC9842108 DOI: 10.1016/j.ijbiomac.2021.10.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 01/19/2023]
Abstract
Ascorbic acid (AA) uptake in neurons occurs via a Na+-dependent carrier-mediated process mediated by the sodium-dependent vitamin C transporter-2 (SVCT2). Relatively little information is available concerning the network of interacting proteins that support human (h)SVCT2 trafficking and cell surface expression in neuronal cells. Here we identified the synaptogenic adhesion protein, calsyntenin-3 (CLSTN3) as an hSVCT2 interacting protein from yeast two-hybrid (Y2H) screening of a human adult brain cDNA library. This interaction was confirmed by co-immunoprecipitation, mammalian two-hybrid (M2H), and co-localization in human cell lines. Co-expression of hCLSTN3 with hSVCT2 in SH-SY5Y cells led to a marked increase in AA uptake. Reciprocally, siRNA targeting hCLSTN3 inhibited AA uptake. In the J20 mouse model of Alzheimer's disease (AD), mouse (m)SVCT2 and mCLSTN3 expression levels in hippocampus were decreased. Similarly, expression levels of hSVCT2 and hCLSTN3 were markedly decreased in hippocampal samples from AD patients. These findings establish CLSTN3 as a novel hSVCT2 interactor in neuronal cells with potential pathophysiological significance.
Collapse
Affiliation(s)
- Veedamali S. Subramanian
- Department of Medicine, University of California, Irvine, CA 92697, United States of America,Corresponding author. (V.S. Subramanian)
| | - Trevor Teafatiller
- Department of Medicine, University of California, Irvine, CA 92697, United States of America
| | - Janielle Vidal
- Department of Medicine, University of California, Irvine, CA 92697, United States of America,Department of Environmental and Occupational Health, University of California, Irvine, CA 92697, United States of America
| | - Gihan S. Gunaratne
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Carlos J. Rodriguez-Ortiz
- Department of Medicine, University of California, Irvine, CA 92697, United States of America,Department of Environmental and Occupational Health, University of California, Irvine, CA 92697, United States of America
| | - Masashi Kitazawa
- Department of Medicine, University of California, Irvine, CA 92697, United States of America,Department of Environmental and Occupational Health, University of California, Irvine, CA 92697, United States of America
| | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| |
Collapse
|
8
|
Sluysmans S, Méan I, Xiao T, Boukhatemi A, Ferreira F, Jond L, Mutero A, Chang CJ, Citi S. PLEKHA5, PLEKHA6, and PLEKHA7 bind to PDZD11 to target the Menkes ATPase ATP7A to the cell periphery and regulate copper homeostasis. Mol Biol Cell 2021; 32:ar34. [PMID: 34613798 PMCID: PMC8693958 DOI: 10.1091/mbc.e21-07-0355] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/12/2023] Open
Abstract
Copper homeostasis is crucial for cellular physiology and development, and its dysregulation leads to disease. The Menkes ATPase ATP7A plays a key role in copper efflux, by trafficking from the Golgi to the plasma membrane upon cell exposure to elevated copper, but the mechanisms that target ATP7A to the cell periphery are poorly understood. PDZD11 interacts with the C-terminus of ATP7A, which contains sequences involved in ATP7A trafficking, but the role of PDZD11 in ATP7A localization is unknown. Here we identify PLEKHA5 and PLEKHA6 as new interactors of PDZD11 that bind to the PDZD11 N-terminus through their WW domains similarly to the junctional protein PLEKHA7. Using CRISPR-KO kidney epithelial cells, we show by immunofluorescence microscopy that WW-PLEKHAs (PLEKHA5, PLEKHA6, PLEKHA7) recruit PDZD11 to distinct plasma membrane localizations and that they are required for the efficient anterograde targeting of ATP7A to the cell periphery in elevated copper conditions. Pull-down experiments show that WW-PLEKHAs promote PDZD11 interaction with the C-terminus of ATP7A. However, WW-PLEKHAs and PDZD11 are not necessary for ATP7A Golgi localization in basal copper, ATP7A copper-induced exit from the Golgi, and ATP7A retrograde trafficking to the Golgi. Finally, measuring bioavailable and total cellular copper, metallothionein-1 expression, and cell viability shows that WW-PLEKHAs and PDZD11 are required for maintaining low intracellular copper levels when cells are exposed to elevated copper. These data indicate that WW-PLEKHAs-PDZD11 complexes regulate the localization and function of ATP7A to promote copper extrusion in elevated copper.
Collapse
Affiliation(s)
- Sophie Sluysmans
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| | - Isabelle Méan
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| | - Tong Xiao
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720
| | - Amina Boukhatemi
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| | - Flavio Ferreira
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| | - Lionel Jond
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| | - Annick Mutero
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Sandra Citi
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1205 Geneva, Switzerland
| |
Collapse
|
9
|
Chen Y, Xie H, Xie T, Yang X, Pang Y, Ye S. Elevated Expression of PDZD11 Is Associated With Poor Prognosis and Immune Infiltrates in Hepatocellular Carcinoma. Front Genet 2021; 12:669928. [PMID: 34093661 PMCID: PMC8176286 DOI: 10.3389/fgene.2021.669928] [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: 02/19/2021] [Accepted: 04/30/2021] [Indexed: 01/11/2023] Open
Abstract
Epithelial cells are held together by tight and adherent junctions, which are destroyed by the activation of epithelial-to-mesenchymal transition (EMT). The PLEKHA7-PDZD11 complex has been reported to be important for epithelial cell adhesion and connecting tissues. However, there is no research regarding the expression and role of PDZD11 in liver hepatocellular carcinoma (LIHC) progression. Here, we analyzed PDZD11 mRNA expression and its clinical results in LIHC patient RNA sequencing data based on different open databases. Furthermore, we examined differences in PDZD11 expression in LIHC tissues and cell lines using western blotting and real-time qPCR. These results are the first to report that the mRNA and protein levels of PDZD11 are significantly overexpressed in LIHC. Moreover, high expression of PDZD11 was correlated with poor overall survival in patients with LIHC. Gene regulatory network analysis suggested that PDZD11 is mainly involved in copper ion homeostasis, proteasome, and oxidative phosphorylation pathways. Interestingly, we found that PDZD11 levels were positively correlated with the abundance of immune infiltrates. In particular, higher infiltration levels of CD4+ T cells and macrophage subsets significantly affected LIHC patient prognosis. Taken together, these results demonstrate that PDZD11 could be a potential diagnostic and prognostic biomarker in LIHC.
Collapse
Affiliation(s)
- Yao Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haifeng Xie
- Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Xie
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xunjun Yang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.,Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yilin Pang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - SongDao Ye
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Identification and targeted management of a neurodegenerative disorder caused by biallelic mutations in SLC5A6. NPJ Genom Med 2019; 4:28. [PMID: 31754459 PMCID: PMC6856110 DOI: 10.1038/s41525-019-0103-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022] Open
Abstract
We describe a sibling pair displaying an early infantile-onset, progressive neurodegenerative phenotype, with symptoms of developmental delay and epileptic encephalopathy developing from 12 to 14 months of age. Using whole exome sequencing, compound heterozygous variants were identified in SLC5A6, which encodes the sodium-dependent multivitamin transporter (SMVT) protein. SMVT is an important transporter of the B-group vitamins biotin, pantothenate, and lipoate. The protein is ubiquitously expressed and has major roles in vitamin uptake in the digestive system, as well as transport of these vitamins across the blood–brain barrier. Pathogenicity of the identified variants was demonstrated by impaired biotin uptake of mutant SMVT. Identification of this vitamin transporter as the genetic basis of this disorder guided targeted therapeutic intervention, resulting clinically in improvement of the patient’s neurocognitive and neuromotor function. This is the second report of biallelic mutations in SLC5A6 leading to a neurodegenerative disorder due to impaired biotin, pantothenate and lipoate uptake. The genetic and phenotypic overlap of these cases confirms mutations in SLC5A6 as the genetic cause of this disease phenotype. Recognition of the genetic disorder caused by SLC5A6 mutations is essential for early diagnosis and to facilitate timely intervention by triple vitamin (biotin, pantothenate, and lipoate) replacement therapy.
Collapse
|
11
|
Sabui S, Subramanian VS, Pham Q, Said HM. Identification of transmembrane protein 237 as a novel interactor with the intestinal riboflavin transporter-3 (RFVT-3): role in functionality and cell biology. Am J Physiol Cell Physiol 2019; 316:C805-C814. [PMID: 30892938 DOI: 10.1152/ajpcell.00029.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The apically localized riboflavin (RF) transporter-3 (RFVT-3) is involved in intestinal absorption of vitamin B2. Previous studies have characterized different physiological/biological aspects of the RFVT-3, but there is a lack of knowledge regarding possible existence of interacting partner(s) and consequence of interaction(s) on its function/cell biology. To address the latter, we performed yeast two-hybrid (Y2H) screening of a human colonic cDNA library and have identified transmembrane protein 237 (TMEM237) as a putative interactor with the human (h)RFVT-3; the interaction was further confirmed via "1-by-1" Y2H assay that involved appropriate positive and negative controls. TMEM237 was found to be highly expressed in human native intestine and in human intestinal epithelial cell lines; further, confocal images showed colocalization of the protein with hRFVT-3. The interaction between TMEM237 with hRFVT-3 in human intestinal epithelial HuTu-80 cells was established by coimmunoprecipitation. Expressing TMEM237 in HuTu-80 cells led to a significant induction in RF uptake, while its knockdown (with the use of gene-specific siRNA) led to a significant reduction in uptake. Transfecting TMEM237 into HuTu-80 cells also led to a marked enhancement in hRFVT-3 protein stability (reflected by an increase in the protein half-life). Interestingly, the level of expression of TMEM237 was found to be markedly reduced following treatment with TNF-α (a proinflammatory cytokine that inhibits intestinal RF uptake), while its expression was significantly upregulated following treatment with butyrate (an inducer of intestinal RF uptake). These findings identify TMEM237 as an interactor with the intestinal hRFVT-3 and show that the interaction has physiological/biological significance.
Collapse
Affiliation(s)
- Subrata Sabui
- Department of Physiology and Biophysics, School of Medicine, University of California , Irvine, California.,Department of Medicine, School of Medicine, University of California , Irvine, California.,Veterans Affairs Medical Center, Long Beach, California
| | - Veedamali S Subramanian
- Department of Physiology and Biophysics, School of Medicine, University of California , Irvine, California.,Department of Medicine, School of Medicine, University of California , Irvine, California.,Veterans Affairs Medical Center, Long Beach, California
| | - Quang Pham
- Department of Physiology and Biophysics, School of Medicine, University of California , Irvine, California
| | - Hamid M Said
- Department of Physiology and Biophysics, School of Medicine, University of California , Irvine, California.,Department of Medicine, School of Medicine, University of California , Irvine, California.,Veterans Affairs Medical Center, Long Beach, California
| |
Collapse
|
12
|
Vasileva E, Sluysmans S, Bochaton-Piallat ML, Citi S. Cell-specific diversity in the expression and organization of cytoplasmic plaque proteins of apical junctions. Ann N Y Acad Sci 2017; 1405:160-176. [PMID: 28617990 DOI: 10.1111/nyas.13391] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 01/11/2023]
Abstract
Tight and adherens junctions play critical roles in the barrier, adhesion, and signaling functions of epithelial and endothelial cells. How the molecular organization of these junctions is tuned to the widely diverse physiological requirements of each tissue type is not well understood. Here, we address this question by examining the expression, localization, and interactions of major cytoplasmic plaque proteins of tight and adherens junctions in different cultured epithelial and endothelial cell lines. Immunoblotting and immunofluorescence analyses show that the expression profiles of cingulin, paracingulin, ZO-1, ZO-2, ZO-3, PLEKHA7, afadin, PDZD11, p120-catenin, and α-catenin, as well as the transmembrane junctional proteins occludin, E-cadherin, and VE-cadherin, are significantly diverse when comparing kidney cells (MDCK, mCCD), keratinocytes (HaCaT), lung carcinoma (A427, A549), and endothelium-derived cells (bEnd.3, meEC, H5V). Proximity ligation and co-immunoprecipitation assays show that PLEKHA7 and PDZD11 are significantly more associated with the tight junction proteins cingulin and ZO-1 in aortic endothelium-derived (meEC) cells but not kidney collecting duct epithelial (mCCD) cells. These results provide evidence that the cytoplasmic plaques of tight and adherens junctions are diverse in their composition and molecular architecture and establish a conceptual framework by which we can rationally address the mechanisms of tissue-dependent junction physiology and signaling by cytoplasmic junctional proteins.
Collapse
Affiliation(s)
- Ekaterina Vasileva
- Department of Cell Biology, Faculty of Sciences, Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Sophie Sluysmans
- Department of Cell Biology, Faculty of Sciences, Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | | | - Sandra Citi
- Department of Cell Biology, Faculty of Sciences, Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| |
Collapse
|
13
|
Guerrera D, Shah J, Vasileva E, Sluysmans S, Méan I, Jond L, Poser I, Mann M, Hyman AA, Citi S. PLEKHA7 Recruits PDZD11 to Adherens Junctions to Stabilize Nectins. J Biol Chem 2016; 291:11016-29. [PMID: 27044745 DOI: 10.1074/jbc.m115.712935] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Indexed: 01/07/2023] Open
Abstract
PLEKHA7 is a junctional protein implicated in stabilization of the cadherin protein complex, hypertension, cardiac contractility, glaucoma, microRNA processing, and susceptibility to bacterial toxins. To gain insight into the molecular basis for the functions of PLEKHA7, we looked for new PLEKHA7 interactors. Here, we report the identification of PDZ domain-containing protein 11 (PDZD11) as a new interactor of PLEKHA7 by yeast two-hybrid screening and by mass spectrometry analysis of PLEKHA7 immunoprecipitates. We show that PDZD11 (17 kDa) is expressed in epithelial and endothelial cells, where it forms a complex with PLEKHA7, as determined by co-immunoprecipitation analysis. The N-terminal Trp-Trp (WW) domain of PLEKHA7 interacts directly with the N-terminal 44 amino acids of PDZD11, as shown by GST-pulldown assays. Immunofluorescence analysis shows that PDZD11 is localized at adherens junctions in a PLEKHA7-dependent manner, because its junctional localization is abolished by knock-out of PLEKHA7, and is rescued by re-expression of exogenous PLEKHA7. The junctional recruitment of nectin-1 and nectin-3 and their protein levels are decreased via proteasome-mediated degradation in epithelial cells where either PDZD11 or PLEKHA7 have been knocked-out. PDZD11 forms a complex with nectin-1 and nectin-3, and its PDZ domain interacts directly with the PDZ-binding motif of nectin-1. PDZD11 is required for the efficient assembly of apical junctions of epithelial cells at early time points in the calcium-switch model. These results show that the PLEKHA7-PDZD11 complex stabilizes nectins to promote efficient early junction assembly and uncover a new molecular mechanism through which PLEKHA7 recruits PDZ-binding membrane proteins to epithelial adherens junctions.
Collapse
Affiliation(s)
- Diego Guerrera
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Jimit Shah
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Ekaterina Vasileva
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Sophie Sluysmans
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Isabelle Méan
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Lionel Jond
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Ina Poser
- the Max Planck Institute for Cell Biology and Genetics, 01307 Dresden, Germany, and
| | - Matthias Mann
- the Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Anthony A Hyman
- the Max Planck Institute for Cell Biology and Genetics, 01307 Dresden, Germany, and
| | - Sandra Citi
- From the Department of Cell Biology and Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland,
| |
Collapse
|
14
|
A systematic evaluation of sorting motifs in the sodium–iodide symporter (NIS). Biochem J 2016; 473:919-28. [DOI: 10.1042/bj20151086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/01/2016] [Indexed: 01/08/2023]
Abstract
Human sodium–iodide symporter (NIS) variants were created to suppress predicted binding motifs potentially implicated in trafficking of this protein. A leucine residue in an internal PDZ-binding motif was found to be essential for expression of the symporter at the plasma membrane.
Collapse
|
15
|
Quick M, Shi L. The sodium/multivitamin transporter: a multipotent system with therapeutic implications. VITAMINS AND HORMONES 2015; 98:63-100. [PMID: 25817866 PMCID: PMC5530880 DOI: 10.1016/bs.vh.2014.12.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The Na(+)/multivitamin transporter (SMVT) is a member of the solute:sodium symporter family that catalyzes the Na(+)-dependent uptake of the structurally diverse water-soluble vitamins pantothenic acid (vitamin B5) and biotin (vitamin H), α-lipoic acid-a vitamin-like substance with strong antioxidant properties-and iodide. The organic substrates of SMVT play central roles in the cellular metabolism and are, therefore, essential for normal human health and development. For example, biotin deficiency leads to growth retardation, dermatological disorders, and neurological disorders. Animal studies have shown that biotin deficiency during pregnancy is directly correlated to embryonic growth retardation, congenital malformation, and death of the embryo. This chapter focuses on the structural and functional features of the human isoform of SMVT (hSMVT); the discovery of which was greatly facilitated by the cloning and expression of hSMVT in tractable expression systems. Special emphasis will be given to mechanistic implications of the transport process of hSMVT that will inform our understanding of the molecular determinants of hSMVT-mediated transport in dynamic context to alleviate the development and optimization of hSMVT as a multipotent platform for drug delivery.
Collapse
Affiliation(s)
- Matthias Quick
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, New York, USA.
| | - Lei Shi
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
16
|
Subramanian VS, Nabokina SM, Said HM. Association of TM4SF4 with the human thiamine transporter-2 in intestinal epithelial cells. Dig Dis Sci 2014; 59:583-90. [PMID: 24282057 PMCID: PMC3943980 DOI: 10.1007/s10620-013-2952-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/12/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND The human thiamine transporter-2 (hTHTR-2) is involved in the intestinal absorption of thiamine. Recent studies with membrane transporters of other nutrients/substrates have shown that they have associated proteins that affect different aspects of their physiology and cell biology. Nothing is known about protein(s) that interact with hTHTR-2 in intestinal epithelial cells and influence its physiological function and/or its cell biology. AIMS The aim of this study was to identify protein partner(s) that interact with hTHTR-2 in human intestinal cells and determine the physiological/biological consequence of that interaction. METHODS The yeast split-ubiquitin two-hybrid approach was used to screen a human intestinal cDNA library. GST-pull-down and cellular co-localization approaches were used to confirm the interaction between hTHTR-2 and the associated protein(s). The effect of such an interaction on hTHTR-2 function was examined by (3)H-thiamine uptake assays. RESULTS Our screening results identified the human TransMembrane 4 SuperFamily 4 (TM4SF4) as a potential interactor with hTHTR-2. This interaction was confirmed by an in vitro GST-pull-down assay, and by live-cell confocal imaging of HuTu-80 cells co-expressing hTHTR-2-GFP and mCherry-TM4SF4 (the latter displayed a significant overlap of these two proteins in intracellular vesicles and at the cell membrane). Co-expression of hTHTR-2 with TM4SF4 in HuTu-80 cells led to a significant induction in thiamine uptake. In contrast, silencing TM4SF4 with gene-specific siRNA led to a significant decrease in thiamine uptake. CONCLUSIONS These results show for the first time that the accessory protein TM4SF4 interacts with hTHTR-2 and influences the physiological function of the thiamine transporter.
Collapse
Affiliation(s)
| | | | - Hamid M. Said
- To whom correspondence may be addressed: , Phone: 562-826-5811; Fax: 562-826-5018
| |
Collapse
|
17
|
Said HM. Recent advances in transport of water-soluble vitamins in organs of the digestive system: a focus on the colon and the pancreas. Am J Physiol Gastrointest Liver Physiol 2013; 305:G601-10. [PMID: 23989008 PMCID: PMC3840235 DOI: 10.1152/ajpgi.00231.2013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This review focuses on recent advances in our understanding of the mechanisms and regulation of water-soluble vitamin (WSV) transport in the large intestine and pancreas, two important organs of the digestive system that have only recently received their fair share of attention. WSV, a group of structurally unrelated compounds, are essential for normal cell function and development and, thus, for overall health and survival of the organism. Humans cannot synthesize WSV endogenously; rather, WSV are obtained from exogenous sources via intestinal absorption. The intestine is exposed to two sources of WSV: a dietary source and a bacterial source (i.e., WSV generated by the large intestinal microbiota). Contribution of the latter source to human nutrition/health has been a subject of debate and doubt, mostly based on the absence of specialized systems for efficient uptake of WSV in the large intestine. However, recent studies utilizing a variety of human and animal colon preparations clearly demonstrate that such systems do exist in the large intestine. This has provided strong support for the idea that the microbiota-generated WSV are of nutritional value to the host, and especially to the nutritional needs of the local colonocytes and their health. In the pancreas, WSV are essential for normal metabolic activities of all its cell types and for its exocrine and endocrine functions. Significant progress has also been made in understanding the mechanisms involved in the uptake of WSV and the effect of chronic alcohol exposure on the uptake processes.
Collapse
Affiliation(s)
- Hamid M. Said
- Departments of Medicine and Physiology/Biophysics, University of California, Irvine, California; and Department of Veterans Affairs Medical Center, Long Beach, California
| |
Collapse
|
18
|
Subramanian VS, Nabokina SM, Patton JR, Marchant JS, Moradi H, Said HM. Glyoxalate reductase/hydroxypyruvate reductase interacts with the sodium-dependent vitamin C transporter-1 to regulate cellular vitamin C homeostasis. Am J Physiol Gastrointest Liver Physiol 2013; 304:G1079-86. [PMID: 23599041 PMCID: PMC3680717 DOI: 10.1152/ajpgi.00090.2013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The human sodium-dependent vitamin C transporter 1 (hSVCT1) contributes to cellular uptake of ascorbic acid (AA). Although different aspects of hSVCT1 cell biology have been extensively studied, nothing is currently known about the broader hSVCT1 interactome that modulates its role in cellular physiology. Here, we identify the enzyme human glyoxalate reductase/hydroxypyruvate reductase (hGR/HPR) as an hSVCT1 associated protein by yeast two-hybrid (Y2H) screening of a human liver cDNA library. The interaction between hSVCT1 and hGR/HPR was further confirmed by in vitro GST pull-down assay, in vivo coimmunoprecipitation and mammalian two-hybrid firefly luciferase assays. This interaction had functional significance as coexpression of hGR/HPR with hSVCT1 led to an increase in AA uptake. Reciprocally, siRNA-mediated knockdown of endogenous hGR/HPR led to an inhibition of AA uptake. Given that oxalate is a degradation product of vitamin C and hGR/HPR acts to limit cellular oxalate levels, this association physically couples two independent regulators of cellular oxalate production. Furthermore, confocal imaging of human liver HepG2 cells coexpressing GFP-hSVCT1 and hGR/HPR-mCherry demonstrated that these two proteins colocalize within a subpopulation of intracellular organelles. This provides a possible molecular basis for organellar AA transport and regulation of local glyoxylate/glycolate concentration in the vicinity of organelle membranes.
Collapse
Affiliation(s)
- Veedamali S. Subramanian
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine, California; ,2Department of Veterans Affairs Medical Center, Long Beach, California; and
| | - Svetlana M. Nabokina
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine, California; ,2Department of Veterans Affairs Medical Center, Long Beach, California; and
| | - Joseph R. Patton
- 3Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jonathan S. Marchant
- 3Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Hamid Moradi
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine, California; ,2Department of Veterans Affairs Medical Center, Long Beach, California; and
| | - Hamid M. Said
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine, California; ,2Department of Veterans Affairs Medical Center, Long Beach, California; and
| |
Collapse
|
19
|
Nabokina SM, Subramanian VS, Said HM. Effect of clinical mutations on functionality of the human riboflavin transporter-2 (hRFT-2). Mol Genet Metab 2012; 105:652-7. [PMID: 22273710 PMCID: PMC3309148 DOI: 10.1016/j.ymgme.2011.12.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 12/27/2011] [Accepted: 12/27/2011] [Indexed: 10/14/2022]
Abstract
The Brown-Vialetto-Van Laere syndrome (BVVLS) is a rare neurological disease characterized by ponto-bulbar palsy, bilateral sensorineural deafness, and respiratory insufficiency. Recent genetic studies have identified mutations in the C20orf54 gene, which encodes the human riboflavin (RF) transporter -2 (hRFT-2) and suggested their link to the manifestation of BVVLS. However, there is nothing currently known about the effect of these mutations on functionality of hRFT-2, a protein that is expressed in a variety of tissues with high expression in the intestine. We addressed this issue using the human-derived intestinal epithelial Caco-2 cells. Our results showed significant (P<0.01) impairment in RF uptake by Caco-2 cells transiently expressing W17R, P28T, E36K, E71K, and R132W (but not L350M) hRFT-2 mutants. This impairment in RF transport was not due to a decrease in transcription and/or translation of hRFT-2, since mRNA and protein levels of the carrier were similar in cells expressing the mutants and wild-type hRFT-2. Confocal images of live Caco-2 cells transiently transfected with hRFT-2 mutants (fused with green fluorescent protein) showed the P28T, E36K, E71K, and R132W mutants were retained within the endoplasmic reticulum, while the W17R and L350M mutants were expressed at the cell membrane; cell surface expression of the W17R mutant was further confirmed by direct determination of cell surface transporter density. These results show for the first time that some of the BVVLS associated mutations in hRFT-2 affect the transporter functionality and that this effect is mediated via alterations in membrane targeting and/or activity of the transporter.
Collapse
Affiliation(s)
- Svetlana M Nabokina
- Departments of Medicine, Physiology/Biophysics, University of California, Irvine, CA 92697, USA
| | | | | |
Collapse
|
20
|
Nabokina SM, Senthilkumar SR, Said HM. Tspan-1 interacts with the thiamine transporter-1 in human intestinal epithelial cells and modulates its stability. Am J Physiol Gastrointest Liver Physiol 2011; 301:G808-13. [PMID: 21836059 PMCID: PMC3220328 DOI: 10.1152/ajpgi.00269.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The human thiamine transporter-1 (hTHTR-1) contributes to intestinal thiamine uptake, and its function is regulated at both the transcriptional and posttranscriptional levels. Nothing, however, is known about the protein(s) that may interact with hTHTR-1 and affects its cell biology and physiology. We addressed this issue in the present investigation using a bacterial two-hybrid system to screen a human intestinal cDNA library with the complete coding sequence of hTHTR-1 as a bait. Our results showed that a member of the tetraspanin family of proteins, Tspan-1, interacts with hTHTR-1. Coimmunoprecipitation and glutathione S-transferase (GST)-pulldown assays confirmed the existence of such an interaction between hTspan-1 and hTHTR-1 in human intestinal epithelial Caco-2 cells. Furthermore, live cell confocal imaging demonstrated that hTspan-1 and hTHTR-1 colocalize in human intestinal epithelial HuTu-80 cells. The importance of the interaction between hTspan-1 and hTHTR-1 for cell biology of the thiamine transporter was examined in HuTu-80 cells stably expressing hTHTR-1. Coexpression of hTspan-1 in these cells led to a significant decrease in the rate of degradation of hTHTR-1 compared with cells expressing the hTHTR-1 alone; in fact the half-life of the hTHTR-1 protein was twice longer in the former cell type compared with the latter cell type (12 h vs. 6 h, respectively). This finding was also confirmed at the functional level when a significantly higher thiamine uptake was observed in cycloheximide-treated (6 h) cells expressing hTHTR-1 together with hTspan-1 compared with those expressing hTHTR-1 alone. These studies demonstrate for the first time that Tspan-1 is an interacting partner with hTHTR-1 and that this interaction affects hTHTR-1 stability.
Collapse
Affiliation(s)
- Svetlana M. Nabokina
- Departments of Medicine, Physiology/Biophysics, University of California, Irvine, California and Department of Veterans Affairs Medical Center, Long Beach, California
| | - Sundar Rajan Senthilkumar
- Departments of Medicine, Physiology/Biophysics, University of California, Irvine, California and Department of Veterans Affairs Medical Center, Long Beach, California
| | - Hamid M. Said
- Departments of Medicine, Physiology/Biophysics, University of California, Irvine, California and Department of Veterans Affairs Medical Center, Long Beach, California
| |
Collapse
|
21
|
Abstract
Our knowledge of the mechanisms and regulation of intestinal absorption of water-soluble vitamins under normal physiological conditions, and of the factors/conditions that affect and interfere with theses processes has been significantly expanded in recent years as a result of the availability of a host of valuable molecular/cellular tools. Although structurally and functionally unrelated, the water-soluble vitamins share the feature of being essential for normal cellular functions, growth and development, and that their deficiency leads to a variety of clinical abnormalities that range from anaemia to growth retardation and neurological disorders. Humans cannot synthesize water-soluble vitamins (with the exception of some endogenous synthesis of niacin) and must obtain these micronutrients from exogenous sources. Thus body homoeostasis of these micronutrients depends on their normal absorption in the intestine. Interference with absorption, which occurs in a variety of conditions (e.g. congenital defects in the digestive or absorptive system, intestinal disease/resection, drug interaction and chronic alcohol use), leads to the development of deficiency (and sub-optimal status) and results in clinical abnormalities. It is well established now that intestinal absorption of the water-soluble vitamins ascorbate, biotin, folate, niacin, pantothenic acid, pyridoxine, riboflavin and thiamin is via specific carrier-mediated processes. These processes are regulated by a variety of factors and conditions, and the regulation involves transcriptional and/or post-transcriptional mechanisms. Also well recognized now is the fact that the large intestine possesses specific and efficient uptake systems to absorb a number of water-soluble vitamins that are synthesized by the normal microflora. This source may contribute to total body vitamin nutrition, and especially towards the cellular nutrition and health of the local colonocytes. The present review aims to outline our current understanding of the mechanisms involved in intestinal absorption of water-soluble vitamins, their regulation, the cell biology of the carriers involved and the factors that negatively affect these absorptive events.
Collapse
Affiliation(s)
- Hamid M Said
- School of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
22
|
Ghosal A, Subramanian VS, Said HM. Role of the putative N-glycosylation and PKC-phosphorylation sites of the human sodium-dependent multivitamin transporter (hSMVT) in function and regulation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:2073-80. [PMID: 21570947 DOI: 10.1016/j.bbamem.2011.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/16/2011] [Accepted: 04/22/2011] [Indexed: 10/18/2022]
Abstract
The sodium-dependent multivitamin transporter (SMVT) is a major biotin transporter in a variety of tissues including the small intestine. The human SMVT (hSMVT) polypeptide is predicted to have four N-glycosylation sites and two putative PKC phosphorylation sites but their role in the function and regulation of the protein is not known and was examined in this investigation. Our results showed that the hSMVT protein is glycosylated and that this glycosylation is important for its function. Studies utilizing site-directed mutagenesis revealed that the N-glycosylation sites at positions Asn(138) and Asn(489) are important for the function of hSMVT and that mutating these sites significantly reduces the V(max) of the biotin uptake process. Mutating the putative PKC phosphorylation site Thr(286) of hSMVT led to a significant decrease in the PMA-induced inhibition in biotin uptake. The latter effect was not mediated via changes in the level of expression of the hSMVT protein and mRNA or in its level of expression at the cell membrane. These findings demonstrate that the hSMVT protein is glycosylated, and that glycosylation is important for its function. Furthermore, the study shows a role for the putative PKC-phosphorylation site Thr(286) of hSMVT in the PKC-mediated regulation of biotin uptake.
Collapse
Affiliation(s)
- Abhisek Ghosal
- Department of Medicine and Physiology/Biophysics, University of California, Irvine, CA 92697, USA
| | | | | |
Collapse
|