1
|
Liu D, Lu N, Zang F, Lu M, Zhang J, Zhao Y, Wan H, Wang M, Li QQ, Wang F, Luo S, Ma M, Shi F, Wu H, Tu J, Zhang Y. Magnetic Resonance Imaging-Based Radiogenomic Analysis Reveals Genomic Determinants for Nanoparticle Delivery into Tumors. ACS NANO 2024; 18:34615-34629. [PMID: 39663893 DOI: 10.1021/acsnano.4c09387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Even though the enhanced permeability and retention (EPR) effect is applicable for the passive targeting of solid tumors, many nanodrugs have failed to achieve meaningful clinical outcomes due to the heterogeneity of EPR effect. Therefore, understanding the mechanism of the EPR effect is crucial to overcome the obstacles nanomedicines face in clinical translation. The aim of this study was to establish a reliable method to increase awareness of the critical influencing factors of nanoparticle (NP) transport into tumors based on the EPR effect using a combined radiogenomics and clinical magnetic resonance imaging (MRI) technique and gene set pathway enrichment analysis. Employing poly(lactic-co-glycolic acid) (PLGA)-coated Fe3O4 NPs as the contrast agent, the monolayer and multilayer distribution of the NPs were observed and quantitatively analyzed by MRI, improving the accuracy of evaluating vascular permeability by MRI. By performing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of genes and pathways, we identified a variety of genes affecting vascular permeability, such as Cldn1, Dlg2, Bves, Prkag3, Cldn10, and Cldn8, which are related to tight junctions and control the permeability of blood vessels in tumors. The method presented here provides an MRI-supported approach to increase the breadth of data collected from genetic screens, reveals genetic evidence of the presence of NPs in tumors and lays a foundation for clinical patient stratification and personalized treatment.
Collapse
Affiliation(s)
- Di Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Na Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fengchao Zang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Mingze Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jingyue Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Hao Wan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Mengjun Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Qian-Qian Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Shouhua Luo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ming Ma
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Haoan Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| |
Collapse
|
2
|
Swan AH, Schindler RFR, Savarese M, Mayer I, Rinné S, Bleser F, Schänzer A, Hahn A, Sabatelli M, Perna F, Chapman K, Pfuhl M, Spivey AC, Decher N, Udd B, Tasca G, Brand T. Differential effects of mutations of POPDC proteins on heteromeric interaction and membrane trafficking. Acta Neuropathol Commun 2023; 11:4. [PMID: 36624536 PMCID: PMC9830914 DOI: 10.1186/s40478-022-01501-w] [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: 11/03/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
The Popeye domain containing (POPDC) genes encode sarcolemma-localized cAMP effector proteins. Mutations in blood vessel epicardial substance (BVES) also known as POPDC1 and POPDC2 have been associated with limb-girdle muscular dystrophy and cardiac arrhythmia. Muscle biopsies of affected patients display impaired membrane trafficking of both POPDC isoforms. Biopsy material of patients carrying mutations in BVES were immunostained with POPDC antibodies. The interaction of POPDC proteins was investigated by co-precipitation, proximity ligation, bioluminescence resonance energy transfer and bimolecular fluorescence complementation. Site-directed mutagenesis was utilised to map the domains involved in protein-protein interaction. Patients carrying a novel homozygous variant, BVES (c.547G > T, p.V183F) displayed only a skeletal muscle pathology and a mild impairment of membrane trafficking of both POPDC isoforms. In contrast, variants such as BVES p.Q153X or POPDC2 p.W188X were associated with a greater impairment of membrane trafficking. Co-transfection analysis in HEK293 cells revealed that POPDC proteins interact with each other through a helix-helix interface located at the C-terminus of the Popeye domain. Site-directed mutagenesis of an array of ultra-conserved hydrophobic residues demonstrated that some of them are required for membrane trafficking of the POPDC1-POPDC2 complex. Mutations in POPDC proteins that cause an impairment in membrane localization affect POPDC complex formation while mutations which leave protein-protein interaction intact likely affect some other essential function of POPDC proteins.
Collapse
Affiliation(s)
- Alexander H. Swan
- grid.7445.20000 0001 2113 8111National Heart and Lung Institute (NHLI), Imperial College London, London, UK ,grid.7445.20000 0001 2113 8111Department of Chemistry, Imperial College London, London, UK
| | - Roland F. R. Schindler
- grid.7445.20000 0001 2113 8111National Heart and Lung Institute (NHLI), Imperial College London, London, UK ,grid.434240.5Present Address: Assay Biology, Domainex Ltd, Cambridge, CB10 1XL UK
| | - Marco Savarese
- grid.7737.40000 0004 0410 2071Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Isabelle Mayer
- grid.7445.20000 0001 2113 8111National Heart and Lung Institute (NHLI), Imperial College London, London, UK
| | - Susanne Rinné
- grid.10253.350000 0004 1936 9756Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Marburg, Germany
| | - Felix Bleser
- grid.10253.350000 0004 1936 9756Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Marburg, Germany
| | - Anne Schänzer
- grid.8664.c0000 0001 2165 8627Institute of Neuropathology, Justus Liebig University Giessen, Giessen, Germany
| | - Andreas Hahn
- grid.8664.c0000 0001 2165 8627Department of Child Neurology, Justus Liebig University Giessen, Giessen, Germany
| | - Mario Sabatelli
- grid.8142.f0000 0001 0941 3192Department of Neurology, Universitá Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Perna
- grid.414603.4Dipartimento Di Scienze Cardiovascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Kathryn Chapman
- grid.434240.5Present Address: Assay Biology, Domainex Ltd, Cambridge, CB10 1XL UK
| | - Mark Pfuhl
- grid.13097.3c0000 0001 2322 6764School of Cardiovascular Medicine and Sciences and Randall Centre, King’s College London, London, UK
| | - Alan C. Spivey
- grid.7445.20000 0001 2113 8111Department of Chemistry, Imperial College London, London, UK
| | - Niels Decher
- grid.8664.c0000 0001 2165 8627Institute of Neuropathology, Justus Liebig University Giessen, Giessen, Germany
| | - Bjarne Udd
- grid.7737.40000 0004 0410 2071Folkhälsan Research Center, University of Helsinki, Helsinki, Finland
| | - Giorgio Tasca
- grid.414603.4Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy ,grid.1006.70000 0001 0462 7212Present Address: John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Thomas Brand
- grid.7445.20000 0001 2113 8111National Heart and Lung Institute (NHLI), Imperial College London, London, UK ,Imperial Centre of Translational and Experimental Medicine, Du Cane Road, London, W120NN UK
| |
Collapse
|
3
|
Baldwin TA, Li Y, Marsden AN, Rinné S, Garza‐Carbajal A, Schindler RFR, Zhang M, Garcia MA, Venna VR, Decher N, Brand T, Dessauer CW. POPDC1 scaffolds a complex of adenylyl cyclase 9 and the potassium channel TREK-1 in heart. EMBO Rep 2022; 23:e55208. [PMID: 36254885 PMCID: PMC9724675 DOI: 10.15252/embr.202255208] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
The establishment of macromolecular complexes by scaffolding proteins is key to the local production of cAMP by anchored adenylyl cyclase (AC) and the subsequent cAMP signaling necessary for cardiac functions. We identify a novel AC scaffold, the Popeye domain-containing (POPDC) protein. The POPDC family of proteins is important for cardiac pacemaking and conduction, due in part to their cAMP-dependent binding and regulation of TREK-1 potassium channels. We show that TREK-1 binds the AC9:POPDC1 complex and copurifies in a POPDC1-dependent manner with AC9 activity in heart. Although the AC9:POPDC1 interaction is cAMP-independent, TREK-1 association with AC9 and POPDC1 is reduced upon stimulation of the β-adrenergic receptor (βAR). AC9 activity is required for βAR reduction of TREK-1 complex formation with AC9:POPDC1 and in reversing POPDC1 enhancement of TREK-1 currents. Finally, deletion of the gene-encoding AC9 (Adcy9) gives rise to bradycardia at rest and stress-induced heart rate variability, a milder phenotype than the loss of Popdc1 but similar to the loss of Kcnk2 (TREK-1). Thus, POPDC1 represents a novel adaptor for AC9 interactions with TREK-1 to regulate heart rate control.
Collapse
Affiliation(s)
- Tanya A Baldwin
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | - Yong Li
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | - Autumn N Marsden
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior MCMBBPhilipps‐University of MarburgMarburgGermany
| | - Anibal Garza‐Carbajal
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | | | - Musi Zhang
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | - Mia A Garcia
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | - Venugopal Reddy Venna
- Department NeurologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior MCMBBPhilipps‐University of MarburgMarburgGermany
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College LondonLondonUK
| | - Carmen W Dessauer
- Department Integrative Biology and PharmacologyMcGovern Medical School, University of Texas Health Science CenterHoustonTXUSA
| |
Collapse
|
4
|
Han P, Lei Y, Liu J, Liu J, Huang H, Tian D, Yan W. Cell adhesion molecule BVES functions as a suppressor of tumor cells extrusion in hepatocellular carcinoma metastasis. Cell Commun Signal 2022; 20:149. [PMID: 36123685 PMCID: PMC9487093 DOI: 10.1186/s12964-022-00962-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background Tumor cells detachment from primary lesions is an early event for hepatocellular carcinoma (HCC) metastasis, in which cell adhesion molecules play an important role. The role of mechanical crowding has attracted increasing attention. Previous studies have found that overcrowding can induce live cells extrusion to maintain epithelial cell homeostasis, and normally, live extruded cells eventually die through a process termed anoikis, suggesting the potential of tumor cells resistant to anoikis might initiate metastasis from primary tumors by cell extrusion. We have demonstrated transmembrane adhesion molecule blood vessel epicardial substance (BVES) suppression as an early event in HCC metastasis. However, whether its suppression is involved in HCC cell extrusion, especially in HCC metastasis, remains unknown. This study aims to investigate the role of BVES in tumor cells extrusion in HCC metastasis, as well as the underlying mechanisms. Methods Cells extrusion was observed by silicone chamber, petri dish inversion, and three-dimensional cell culture model. Polymerase chain reaction, western blotting, immunohistochemistry, immunofluorescence, co-immunoprecipitation, and RhoA activity assays were used to explore the underlying mechanisms of cell extrusion regulated by BVES. An orthotopic xenograft model was established to investigate the effects of BVES and cell extrusion in HCC metastasis in vivo. Results Tumor cell extrusion was observed in HCC cells and tissues. BVES expression was decreased both in HCC and extruded tumor cells. BVES overexpression led to the decrease in HCC cells extrusion in vitro and in vivo. Moreover, our data showed that BVES co-localized with ZO-1 and GEFT, regulating ZO-1 expression and localization, and GEFT distribution, thus modulating RhoA activity. Conclusion The present study revealed that BVES downregulation in HCC enhanced tumor cells extrusion, thus promoting HCC metastasis, which contributed to a more comprehensive understanding of tumor metastasis, and provided clues for developing novel HCC therapy strategies. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00962-9.
Collapse
Affiliation(s)
- Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Jingmei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Jiqiao Liu
- Department of Ultrasound, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Huanjun Huang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
5
|
Ahmed MB, Alghamdi AAA, Islam SU, Lee JS, Lee YS. cAMP Signaling in Cancer: A PKA-CREB and EPAC-Centric Approach. Cells 2022; 11:cells11132020. [PMID: 35805104 PMCID: PMC9266045 DOI: 10.3390/cells11132020] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is one of the most common causes of death globally. Despite extensive research and considerable advances in cancer therapy, the fundamentals of the disease remain unclear. Understanding the key signaling mechanisms that cause cancer cell malignancy may help to uncover new pharmaco-targets. Cyclic adenosine monophosphate (cAMP) regulates various biological functions, including those in malignant cells. Understanding intracellular second messenger pathways is crucial for identifying downstream proteins involved in cancer growth and development. cAMP regulates cell signaling and a variety of physiological and pathological activities. There may be an impact on gene transcription from protein kinase A (PKA) as well as its downstream effectors, such as cAMP response element-binding protein (CREB). The position of CREB downstream of numerous growth signaling pathways implies its oncogenic potential in tumor cells. Tumor growth is associated with increased CREB expression and activation. PKA can be used as both an onco-drug target and a biomarker to find, identify, and stage tumors. Exploring cAMP effectors and their downstream pathways in cancer has become easier using exchange protein directly activated by cAMP (EPAC) modulators. This signaling system may inhibit or accelerate tumor growth depending on the tumor and its environment. As cAMP and its effectors are critical for cancer development, targeting them may be a useful cancer treatment strategy. Moreover, by reviewing the material from a distinct viewpoint, this review aims to give a knowledge of the impact of the cAMP signaling pathway and the related effectors on cancer incidence and development. These innovative insights seek to encourage the development of novel treatment techniques and new approaches.
Collapse
Affiliation(s)
- Muhammad Bilal Ahmed
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | | | - Salman Ul Islam
- Department of Pharmacy, Cecos University, Peshawar, Street 1, Sector F 5 Phase 6 Hayatabad, Peshawar 25000, Pakistan;
| | - Joon-Seok Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | - Young-Sup Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
6
|
Gruscheski L, Brand T. The Role of POPDC Proteins in Cardiac Pacemaking and Conduction. J Cardiovasc Dev Dis 2021; 8:160. [PMID: 34940515 PMCID: PMC8706714 DOI: 10.3390/jcdd8120160] [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: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
The Popeye domain-containing (POPDC) gene family, consisting of Popdc1 (also known as Bves), Popdc2, and Popdc3, encodes transmembrane proteins abundantly expressed in striated muscle. POPDC proteins have recently been identified as cAMP effector proteins and have been proposed to be part of the protein network involved in cAMP signaling. However, their exact biochemical activity is presently poorly understood. Loss-of-function mutations in animal models causes abnormalities in skeletal muscle regeneration, conduction, and heart rate adaptation after stress. Likewise, patients carrying missense or nonsense mutations in POPDC genes have been associated with cardiac arrhythmias and limb-girdle muscular dystrophy. In this review, we introduce the POPDC protein family, and describe their structure function, and role in cAMP signaling. Furthermore, the pathological phenotypes observed in zebrafish and mouse models and the clinical and molecular pathologies in patients carrying POPDC mutations are described.
Collapse
Affiliation(s)
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
7
|
Tucker SJ, Zorn AJ. The role of Popeye domain-containing protein 1 (POPDC1) in the progression of the malignant phenotype. Br J Pharmacol 2021; 179:2829-2843. [PMID: 33533478 DOI: 10.1111/bph.15403] [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: 09/22/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/14/2022] Open
Abstract
The Popeye domain-containing protein 1 (POPDC1), a tight junction-associated transmembrane protein with a unique binding site for cAMP, has been shown to act as a tumour suppressor in cancer cells. Through interaction with many downstream effectors and signalling pathways, POPDC1 promotes cell adhesion and inhibits uncontrolled cell proliferation, epithelial-to-mesenchymal transition and metastasis. However, POPDC1 expression is down-regulated in many types of cancer, thereby reducing its tumour-suppressive actions. This review discusses the role of POPDC1 in the progression of the malignant phenotype and highlights the broad range of benefits POPDC1 stabilisation may achieve therapeutically. Cancer stem cells (CSCs) are a key hallmark of malignancies and commonly promote treatment resistance. This article provides a comprehensive overview of CSC signalling mechanisms, many of which have been shown to be regulated by POPDC1 in other cell types, thus suggesting an additional therapeutic benefit for POPDC1-stabilising anti-cancer drugs.
Collapse
Affiliation(s)
- Steven J Tucker
- School of Medicine, Medical Science and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Alina J Zorn
- School of Medicine, Medical Science and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
8
|
POPDC proteins and cardiac function. Biochem Soc Trans 2020; 47:1393-1404. [PMID: 31551355 DOI: 10.1042/bst20190249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 01/01/2023]
Abstract
The Popeye domain-containing gene family encodes a novel class of cAMP effector proteins in striated muscle tissue. In this short review, we first introduce the protein family and discuss their structure and function with an emphasis on their role in cyclic AMP signalling. Another focus of this review is the recently discovered role of POPDC genes as striated muscle disease genes, which have been associated with cardiac arrhythmia and muscular dystrophy. The pathological phenotypes observed in patients will be compared with phenotypes present in null and knockin mutations in zebrafish and mouse. A number of protein-protein interaction partners have been discovered and the potential role of POPDC proteins to control the subcellular localization and function of these interacting proteins will be discussed. Finally, we outline several areas, where research is urgently needed.
Collapse
|
9
|
The Role of the Popeye Domain Containing Gene Family in Organ Homeostasis. Cells 2019; 8:cells8121594. [PMID: 31817925 PMCID: PMC6952887 DOI: 10.3390/cells8121594] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The Popeye domain containing (POPDC) gene family consists of POPDC1 (also known as BVES), POPDC2 and POPDC3 and encodes a novel class of cyclic adenosine monophosphate (cAMP) effector proteins. Despite first reports of their isolation and initial characterization at the protein level dating back 20 years, only recently major advances in defining their biological functions and disease association have been made. Loss-of-function experiments in mice and zebrafish established an important role in skeletal muscle regeneration, heart rhythm control and stress signaling. Patients suffering from muscular dystrophy and atrioventricular block were found to carry missense and nonsense mutations in either of the three POPDC genes, which suggests an important function in the control of striated muscle homeostasis. However, POPDC genes are also expressed in a number of epithelial cells and function as tumor suppressor genes involved in the control of epithelial structure, tight junction formation and signaling. Suppression of POPDC genes enhances tumor cell proliferation, migration, invasion and metastasis in a variety of human cancers, thus promoting a malignant phenotype. Moreover, downregulation of POPDC1 and POPDC3 expression in different cancer types has been associated with poor prognosis. However, high POPDC3 expression has also been correlated to poor clinical prognosis in head and neck squamous cell carcinoma, suggesting that POPDC3 potentially plays different roles in the progression of different types of cancer. Interestingly, a gain of POPDC1 function in tumor cells inhibits cell proliferation, migration and invasion thereby reducing malignancy. Furthermore, POPDC proteins have been implicated in the control of cell cycle genes and epidermal growth factor and Wnt signaling. Work in tumor cell lines suggest that cyclic nucleotide binding may also be important in epithelial cells. Thus, POPDC proteins have a prominent role in tissue homeostasis and cellular signaling in both epithelia and striated muscle.
Collapse
|
10
|
Heinemann U, Schuetz A. Structural Features of Tight-Junction Proteins. Int J Mol Sci 2019; 20:E6020. [PMID: 31795346 PMCID: PMC6928914 DOI: 10.3390/ijms20236020] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
Tight junctions are complex supramolecular entities composed of integral membrane proteins, membrane-associated and soluble cytoplasmic proteins engaging in an intricate and dynamic system of protein-protein interactions. Three-dimensional structures of several tight-junction proteins or their isolated domains have been determined by X-ray crystallography, nuclear magnetic resonance spectroscopy, and cryo-electron microscopy. These structures provide direct insight into molecular interactions that contribute to the formation, integrity, or function of tight junctions. In addition, the known experimental structures have allowed the modeling of ligand-binding events involving tight-junction proteins. Here, we review the published structures of tight-junction proteins. We show that these proteins are composed of a limited set of structural motifs and highlight common types of interactions between tight-junction proteins and their ligands involving these motifs.
Collapse
Affiliation(s)
- Udo Heinemann
- Macromolecular Structure and Interaction Laboratory, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Anja Schuetz
- Protein Production & Characterization Platform, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
11
|
Thompson JJ, Short SP, Parang B, Brown RE, Li C, Ng VH, Saito-Diaz K, Choksi YA, Washington MK, Smith JJ, Fingleton B, Brand T, Lee E, Coffey RJ, Williams CS. Blood vessel epicardial substance reduces LRP6 receptor and cytoplasmic β-catenin levels to modulate Wnt signaling and intestinal homeostasis. Carcinogenesis 2019; 40:1086-1098. [PMID: 30689807 PMCID: PMC8067673 DOI: 10.1093/carcin/bgz007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/19/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022] Open
Abstract
Blood vessel epicardial substance (BVES, otherwise known as POPDC1) is an integral membrane protein known to regulate tight junction formation and epithelial-mesenchymal transition. BVES is underexpressed in a number of malignancies, including colorectal cancer. BVES loss leads to activation of the Wnt pathway, suggesting that decreased BVES expression functionally contributes to tumorigenesis. However, the mechanism by which BVES modulates Wnt signaling is unknown. Here, we confirm that BVES loss increases β-catenin protein levels, leads to Wnt pathway activation in a ligand-independent fashion and coordinates with Wnt ligand to further increase Wnt signaling. We show that BVES loss increases levels and activation of the Wnt co-receptor, LRP6, in cell lines, murine adenoma tumoroids and human-derived colonoids. We also demonstrate that BVES interacts with LRP6. Finally, murine tumor modeling using a Wnt-driven genetic model and a chemically induced model of colorectal carcinogenesis demonstrate that BVES loss increases tumor multiplicity and dysplasia. Together, these results implicate BVES as an inhibitor of Wnt signaling, provide one of the first examples of a tight junction-associated protein regulating Wnt receptor levels, and expand the number of putative molecular targets for therapeutic intervention in colorectal cancer.
Collapse
Affiliation(s)
- Joshua J Thompson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sarah P Short
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Bobak Parang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rachel E Brown
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Chenxuan Li
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Victoria H Ng
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kenyi Saito-Diaz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yash A Choksi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mary K Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jesse Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barbara Fingleton
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ethan Lee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Health Care System, Nashville, TN, USA
| | - Christopher S Williams
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Health Care System, Nashville, TN, USA
| |
Collapse
|
12
|
Swan AH, Gruscheski L, Boland LA, Brand T. The Popeye domain containing gene family encoding a family of cAMP-effector proteins with important functions in striated muscle and beyond. J Muscle Res Cell Motil 2019; 40:169-183. [PMID: 31197601 PMCID: PMC6726836 DOI: 10.1007/s10974-019-09523-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
The Popeye domain containing (POPDC) gene family encodes a novel class of membrane-bound cyclic AMP effector proteins. POPDC proteins are abundantly expressed in cardiac and skeletal muscle. Consistent with its predominant expression in striated muscle, Popdc1 and Popdc2 null mutants in mouse and zebrafish develop cardiac arrhythmia and muscular dystrophy. Likewise, mutations in POPDC genes in patients have been associated with cardiac arrhythmia and muscular dystrophy phenotypes. A membrane trafficking function has been identified in this context. POPDC proteins have also been linked to tumour formation. Here, POPDC1 plays a role as a tumour suppressor by limiting c-Myc and WNT signalling. Currently, a common functional link between POPDC's role in striated muscle and as a tumour suppressor is lacking. We also discuss several alternative working models to better understand POPDC protein function.
Collapse
Affiliation(s)
- Alexander H Swan
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
- Institute of Chemical Biology, Imperial College London, London, UK
| | - Lena Gruscheski
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
| | - Lauren A Boland
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
13
|
Han P, Lei Y, Li D, Liu J, Yan W, Tian D. Ten years of research on the role of BVES/ POPDC1 in human disease: a review. Onco Targets Ther 2019; 12:1279-1291. [PMID: 30863095 PMCID: PMC6388986 DOI: 10.2147/ott.s192364] [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] [Indexed: 01/01/2023] Open
Abstract
Since the blood vessel epicardial substance or Popeye domain-containing protein 1 (BVES/POPDC1) was first identified in the developing heart by two independent laboratories in 1999, an increasing number of studies have investigated the structure, function, and related diseases of BVES/POPDC1. During the first 10 years following the discovery of BVES/POPDC1, studies focused mainly on its structure, expression patterns, and functions. Based on these studies, further investigations conducted over the previous decade examined the role of BVES/POPDC1 in human diseases, such as colitis, heart diseases, and human cancers. This review provides an overview of the structure and expression of BVES/POPDC1, mainly focusing on its potential role and mechanism through which it is involved in human cancers.
Collapse
Affiliation(s)
- Ping Han
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, ;
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, ;
| | - Dongxiao Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, ;
| | - Jingmei Liu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, ;
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, ;
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China, ;
| |
Collapse
|
14
|
Parang B, Thompson JJ, Williams CS. Blood Vessel Epicardial Substance (BVES) in junctional signaling and cancer. Tissue Barriers 2018; 6:1-12. [PMID: 30307367 DOI: 10.1080/21688370.2018.1499843] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Blood vessel epicardial substance (BVES) is a tight-junction associated protein that was originally discovered from a cDNA screen of the developing heart. Research over the last decade has shown that not only is BVES is expressed in cardiac and skeletal tissue, but BVES is also is expressed throughout the gastrointestinal epithelium. Mice lacking BVES sustain worse intestinal injury and inflammation. Furthermore, BVES is suppressed in gastrointestinal cancers, and mouse modeling has shown that loss of BVES promotes tumor formation. Recent work from multiple laboratories has revealed that BVES can regulate several molecular pathways, including cAMP, WNT, and promoting the degradation of the oncogene, c-Myc. This review will summarize our current understanding of how BVES regulates the intestinal epithelium and discuss how BVES functions at the molecular level to preserve epithelial phenotypes and suppress tumorigenesis.
Collapse
Affiliation(s)
- Bobak Parang
- a Department of Medicine , Cornell University , New York , NY , USA
| | - Joshua J Thompson
- b Department of Medicine, Division of Gastroenterology , Vanderbilt University , Nashville , TN , USA
| | - Christopher S Williams
- b Department of Medicine, Division of Gastroenterology , Vanderbilt University , Nashville , TN , USA.,c Veterans Affairs Tennessee Valley Health Care System , Nashville , TN , USA
| |
Collapse
|
15
|
Abstract
The Popeye domain containing (POPDC) genes encode transmembrane proteins, which are abundantly expressed in striated muscle cells. Hallmarks of the POPDC proteins are the presence of three transmembrane domains and the Popeye domain, which makes up a large part of the cytoplasmic portion of the protein and functions as a cAMP-binding domain. Interestingly, despite the prediction of structural similarity between the Popeye domain and other cAMP binding domains, at the protein sequence level they strongly differ from each other suggesting an independent evolutionary origin of POPDC proteins. Loss-of-function experiments in zebrafish and mouse established an important role of POPDC proteins for cardiac conduction and heart rate adaptation after stress. Loss-of function mutations in patients have been associated with limb-girdle muscular dystrophy and AV-block. These data suggest an important role of these proteins in the maintenance of structure and function of striated muscle cells.
Collapse
|
16
|
Hou A, Fu J, Shi Y, Qiao L, Li J, Xing Y, Xue X. Decreased ZONAB expression promotes excessive transdifferentiation of alveolar epithelial cells in hyperoxia-induced bronchopulmonary dysplasia. Int J Mol Med 2018; 41:2339-2349. [PMID: 29393348 DOI: 10.3892/ijmm.2018.3413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 01/10/2018] [Indexed: 11/06/2022] Open
Abstract
Previous studies by our group have confirmed excessive transdifferentiation of alveolar epithelial cells (AECs) in a hyperoxia‑induced bronchopulmonary dysplasia (BPD) model, but the underlying mechanism have remained elusive. The transcription factor zonula occludens 1‑associated nucleic acid binding protein (ZONAB) has the biological functions of inhibition of epithelial cell differentiation and promotion of epithelial cell proliferation. The aim of the present study was to explore the regulatory effect of ZONAB on the transdifferentiation and proliferation of AECs in a model of hyperoxia‑induced lung injury. Newborn Wistar rats were randomly allocated to a model group (inhalation of 85% O2) or a control group (inhalation of normal air), and ZONAB expression in lung tissues was detected at different time‑points. Type II AECs (AEC II) isolated from normal newborn rats were primarily cultured under an atmosphere of 85 or 21% O2, and ZONAB expression in the cells was examined. The primary cells were further transfected with ZONAB plasmid or small interfering (si)RNA and then exposed to hyperoxia, and the indicators for transdifferentiation and proliferation were measured. The present study indicated that ZONAB expression in AEC II of the BPD rats was significantly decreased from 7 days of exposure to hyperoxia onwards. In the AEC II isolated from normal neonatal rats, ZONAB expression in the model group was also reduced compared with that in the control group. After transfection with the plasmid pCMV6‑ZONAB, the expression of aquaporin 5 (type I alveolar epithelial cell marker) decreased and the expression of surfactant protein C (AEC II marker), proliferating‑cell nuclear antigen and cyclin D1 increased, which was opposite to the effects of ZONAB siRNA. Transfection with pCMV6‑ZONAB also alleviated excessive transdifferentiation and inhibited proliferation of AEC II induced by hyperoxia treatment. These results suggest that ZONAB expression in AEC II decreases under hyperoxia conditions, which promotes transdifferentiation and inhibits proliferation of AECs. This may, at least in part, be the underlying mechanism of lung epithelial injury in the hyperoxia-induced BPD model.
Collapse
Affiliation(s)
- Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Lin Qiao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jun Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yujiao Xing
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
17
|
Dysregulation of POPDC1 promotes breast cancer cell migration and proliferation. Biosci Rep 2017; 37:BSR20171039. [PMID: 28954821 PMCID: PMC5696453 DOI: 10.1042/bsr20171039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 11/22/2022] Open
Abstract
Breast cancer subtypes such as triple-negative that lack the expression of oestrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor 2 receptor (HER2), remain poorly clinically managed due to a lack of therapeutic targets. This necessitates identification and validation of novel targets. Suppression of Popeye domain-containing protein 1 (POPDC1) is known to promote tumorigenesis and correlate to poor clinical outcomes in various cancers, and also promotes cardiac and skeletal muscle pathologies. It remains to be established whether POPDC1 is dysregulated in breast cancer, and whether overcoming the dysregulation of POPDC1 could present a potential therapeutic strategy to inhibit breast tumorigenesis. We assessed the potential of POPDC1 as a novel target for inhibiting breast cancer cell migration and proliferation. POPDC1 was significantly suppressed with reduced cell membrane localization in breast cancer cells. Furthermore, functional suppression of POPDC1 promoted breast cancer cell migration and proliferation, which were inhibited by POPDC1 overexpression. Finally, cAMP interacts with POPDC1 and up-regulates its expression in breast cancer cells. These findings suggest that POPDC1 plays a role in breast tumorigenesis and represents a potential therapeutic target or biomarker in breast cancer medicine.
Collapse
|
18
|
Amunjela JN, Tucker SJ. POPDC proteins as potential novel therapeutic targets in cancer. Drug Discov Today 2016; 21:1920-1927. [PMID: 27458118 DOI: 10.1016/j.drudis.2016.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/10/2016] [Accepted: 07/18/2016] [Indexed: 02/08/2023]
Abstract
Popeye domain-containing (POPDC) proteins are a novel class of cAMP-binding molecules that affect cancer cell behaviour and correlate with poor clinical outcomes. They are encoded by the POPDC genes POPDC1, POPDC2, and POPDC3. The deletion of POPDC genes and the suppression of POPDC proteins correlate with enhanced cancer cell proliferation, migration, invasion, metastasis, drug resistance, and poor patient survival in various human cancers. Overexpression of POPDC proteins inhibits cancer cell migration and invasion in vitro. POPDC proteins present promising anticancer therapeutic targets and here we review their roles in promoting cancer progression and highlight their potential as anticancer therapeutic targets.
Collapse
Affiliation(s)
- Johanna N Amunjela
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Steven J Tucker
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
19
|
Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol 2016; 17:564-80. [PMID: 27353478 DOI: 10.1038/nrm.2016.80] [Citation(s) in RCA: 994] [Impact Index Per Article: 110.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epithelia and endothelia separate different tissue compartments and protect multicellular organisms from the outside world. This requires the formation of tight junctions, selective gates that control paracellular diffusion of ions and solutes. Tight junctions also form the border between the apical and basolateral plasma-membrane domains and are linked to the machinery that controls apicobasal polarization. Additionally, signalling networks that guide diverse cell behaviours and functions are connected to tight junctions, transmitting information to and from the cytoskeleton, nucleus and different cell adhesion complexes. Recent advances have broadened our understanding of the molecular architecture and cellular functions of tight junctions.
Collapse
Affiliation(s)
- Ceniz Zihni
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Clare Mills
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Karl Matter
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Maria S Balda
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| |
Collapse
|
20
|
Zhu C, Sauter E, Schreiter A, van Roeyen CRC, Ostendorf T, Floege J, Gembardt F, Hugo CP, Isermann B, Lindquist JA, Mertens PR. Cold Shock Proteins Mediate GN with Mesangioproliferation. J Am Soc Nephrol 2016; 27:3678-3689. [PMID: 27151923 DOI: 10.1681/asn.2015121367] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/09/2016] [Indexed: 11/03/2022] Open
Abstract
DNA binding protein A (DbpA) is a member of the human cold shock domain-containing protein superfamily, with known functions in cell proliferation, differentiation, and stress responses. DbpA mediates tight junction-associated activities in tubular epithelial cells, but the function of DbpA in mesangial cells is unknown. Here, we found DbpA protein expression restricted to vascular smooth muscle cells in healthy human kidney tissue but profound induction of DbpA protein expression within the glomerular mesangial compartment in mesangioproliferative nephritis. In vitro, depletion or overexpression of DbpA using lentiviral constructs led to inhibition or promotion, respectively, of mesangial cell proliferation. Because platelet-derived growth factor B (PDGF-B) signaling has a pivotal role in mesangial cell proliferation, we examined the regulatory effect of PDGF-B on DbpA. In vitro studies of human and rat mesangial cells confirmed a stimulatory effect of PDGF-B on DbpA transcript numbers and protein levels. Additional in vivo investigations showed DbpA upregulation in experimental rat anti-Thy1.1 nephritis and murine mesangioproliferative nephritis models. To interfere with PDGF-B signaling, we injected nephritic rats with PDGF-B neutralizing aptamers or the MEK/ERK inhibitor U0126. Both interventions markedly decreased DbpA protein expression. Conversely, continuous PDGF-B infusion in healthy rats induced DbpA expression predominantly within the mesangial compartment. Taken together, these results indicate that DbpA is a novel target of PDGF-B signaling and a key mediator of mesangial cell proliferation.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| | - Eva Sauter
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| | - Anja Schreiter
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| | - Claudia R C van Roeyen
- Department of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; and
| | - Tammo Ostendorf
- Department of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; and
| | - Jürgen Floege
- Department of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; and
| | - Florian Gembardt
- Division of Nephrology, Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Christian P Hugo
- Division of Nephrology, Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | | | - Peter R Mertens
- Department of Nephrology, Hypertension, Diabetes and Endocrinology and
| |
Collapse
|
21
|
Reddy VK, Short SP, Barrett CW, Mittal MK, Keating CE, Thompson JJ, Harris EI, Revetta F, Bader DM, Brand T, Washington MK, Williams CS. BVES Regulates Intestinal Stem Cell Programs and Intestinal Crypt Viability after Radiation. Stem Cells 2016; 34:1626-36. [PMID: 26891025 DOI: 10.1002/stem.2307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 12/24/2015] [Indexed: 01/17/2023]
Abstract
Blood vessel epicardial substance (BVES/Popdc1) is a junctional-associated transmembrane protein that is underexpressed in a number of malignancies and regulates epithelial-to-mesenchymal transition. We previously identified a role for BVES in regulation of the Wnt pathway, a modulator of intestinal stem cell programs, but its role in small intestinal (SI) biology remains unexplored. We hypothesized that BVES influences intestinal stem cell programs and is critical to SI homeostasis after radiation injury. At baseline, Bves(-/-) mice demonstrated increased crypt height, as well as elevated proliferation and expression of the stem cell marker Lgr5 compared to wild-type (WT) mice. Intercross with Lgr5-EGFP reporter mice confirmed expansion of the stem cell compartment in Bves(-/-) mice. To examine stem cell function after BVES deletion, we used ex vivo 3D-enteroid cultures. Bves(-/-) enteroids demonstrated increased stemness compared to WT, when examining parameters such as plating efficiency, stem spheroid formation, and retention of peripheral cystic structures. Furthermore, we observed increased proliferation, expression of crypt-base columnar "CBC" and "+4" stem cell markers, amplified Wnt signaling, and responsiveness to Wnt activation in the Bves(-/-) enteroids. Bves expression was downregulated after radiation in WT mice. Moreover, after radiation, Bves(-/-) mice demonstrated significantly greater SI crypt viability, proliferation, and amplified Wnt signaling in comparison to WT mice. Bves(-/-) mice also demonstrated elevations in Lgr5 and Ascl2 expression, and putative damage-responsive stem cell populations marked by Bmi1 and TERT. Therefore, BVES is a key regulator of intestinal stem cell programs and mucosal homeostasis. Stem Cells 2016;34:1626-1636.
Collapse
Affiliation(s)
- Vishruth K Reddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sarah P Short
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Caitlyn W Barrett
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mukul K Mittal
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cody E Keating
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joshua J Thompson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elizabeth I Harris
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Frank Revetta
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David M Bader
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Thomas Brand
- Developmental Dynamics, Heart Science Centre, Imperial College London, London, U.K
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher S Williams
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA
| |
Collapse
|
22
|
Schindler RFR, Brand T. The Popeye domain containing protein family--A novel class of cAMP effectors with important functions in multiple tissues. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:28-36. [PMID: 26772438 PMCID: PMC4821176 DOI: 10.1016/j.pbiomolbio.2016.01.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/03/2015] [Accepted: 01/04/2016] [Indexed: 12/12/2022]
Abstract
Popeye domain containing (Popdc) proteins are a unique family, which combine several different properties and functions in a surprisingly complex fashion. They are expressed in multiple tissues and cell types, present in several subcellular compartments, interact with different classes of proteins, and are associated with a variety of physiological and pathophysiological processes. Moreover, Popdc proteins bind the second messenger cAMP with high affinity and it is thought that they act as a novel class of cAMP effector proteins. Here, we will review the most important findings about the Popdc family, which accumulated since its discovery about 15 years ago. We will be focussing on Popdc protein interaction and function in striated muscle tissue. However, as a full picture only emerges if all aspects are taken into account, we will also describe what is currently known about the role of Popdc proteins in epithelial cells and in various types of cancer, and discuss these findings with regard to their relevance for cardiac and skeletal muscle.
Collapse
Affiliation(s)
- Roland F R Schindler
- Heart Science Centre, National Heart and Lung Institute (NHLI), Imperial College London, United Kingdom
| | - Thomas Brand
- Heart Science Centre, National Heart and Lung Institute (NHLI), Imperial College London, United Kingdom.
| |
Collapse
|
23
|
Zihni C, Balda MS, Matter K. Signalling at tight junctions during epithelial differentiation and microbial pathogenesis. J Cell Sci 2015; 127:3401-13. [PMID: 25125573 DOI: 10.1242/jcs.145029] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tight junctions are a component of the epithelial junctional complex, and they form the paracellular diffusion barrier that enables epithelial cells to create cellular sheets that separate compartments with different compositions. The assembly and function of tight junctions are intimately linked to the actomyosin cytoskeleton and, hence, are under the control of signalling mechanisms that regulate cytoskeletal dynamics. Tight junctions not only receive signals that guide their assembly and function, but transmit information to the cell interior to regulate cell proliferation, migration and survival. As a crucial component of the epithelial barrier, they are often targeted by pathogenic viruses and bacteria, aiding infection and the development of disease. In this Commentary, we review recent progress in the understanding of the molecular signalling mechanisms that drive junction assembly and function, and the signalling processes by which tight junctions regulate cell behaviour and survival. We also discuss the way in which junctional components are exploited by pathogenic viruses and bacteria, and how this might affect junctional signalling mechanisms.
Collapse
Affiliation(s)
- Ceniz Zihni
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Maria S Balda
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Karl Matter
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| |
Collapse
|
24
|
RhoGTPase signalling at epithelial tight junctions: Bridging the GAP between polarity and cancer. Int J Biochem Cell Biol 2015; 64:120-5. [PMID: 25757376 PMCID: PMC4503795 DOI: 10.1016/j.biocel.2015.02.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/17/2015] [Accepted: 02/26/2015] [Indexed: 01/19/2023]
Abstract
The establishment and maintenance of epithelial polarity must be correctly controlled for normal development and homeostasis. Tight junctions (TJ) in vertebrates define apical and basolateral membrane domains in polarized epithelia via bi-directional, complex signalling pathways between TJ themselves and the cytoskeleton they are associated with. RhoGTPases are central to these processes and evidence suggests that their regulation is coordinated by interactions between GEFs and GAPs with junctional, cytoplasmic adapter proteins. In this InFocus review we determine that the expression, localization or stability of a variety of these adaptor proteins is altered in various cancers, potentially representing an important mechanistic link between loss of polarity and cancer. We focus here, on two well characterized RhoGTPases Cdc42 and RhoA who's GEFs and GAPs are predominantly localized to TJ via cytoplasmic adaptor proteins.
Collapse
|
25
|
Knockdown of zebrafish blood vessel epicardial substance results in incomplete retinal lamination. ScientificWorldJournal 2014; 2014:803718. [PMID: 24741362 PMCID: PMC3972863 DOI: 10.1155/2014/803718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/29/2014] [Indexed: 01/21/2023] Open
Abstract
Cell polarity during eye development determines the normal retinal lamination and differentiation of photoreceptor cells in the retina. In vertebrates, blood vessel epicardial substance (Bves) is known to play an important role in the formation and maintenance of the tight junctions essential for epithelial cell polarity. In the current study, we generated a transgenic zebrafish Bves (zbves) promoter-EGFP zebrafish line to investigate the expression pattern of Bves in the retina and to study the role of zbves in retinal lamination. Immunostaining with different specific antibodies from retinal cells and transmission electron microscopy were used to identify the morphological defects in normal and Bves knockdown zebrafish. In normal zebrafish, Bves is located at the apical junctions of embryonic retinal neuroepithelia during retinogenesis; later, it is strongly expressed around inner plexiform layer (IPL) and retinal pigment epithelium (RPE). In contrast, a loss of normal retinal lamination and cellular polarity was found with undifferentiated photoreceptor cells in Bves knockdown zebrafish. Herein, our results indicated that disruption of Bves will result in a loss of normal retinal lamination.
Collapse
|
26
|
Szaszi K, Amoozadeh Y. New Insights into Functions, Regulation, and Pathological Roles of Tight Junctions in Kidney Tubular Epithelium. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 308:205-71. [DOI: 10.1016/b978-0-12-800097-7.00006-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
27
|
Abstract
Epithelial transport relies on the proper function and regulation of the tight junction (TJ), other-wise uncontrolled paracellular leakage of solutes and water would occur. They also act as a fence against mixing of membrane proteins of the apical and basolateral side. The proteins determining paracellular transport consist of four transmembrane regions, intracellular N and C terminals, one intracellular and two extracellular loops (ECLs). The ECLs interact laterally and with counterparts of the neighboring cell and by this achieve a general sealing function. Two TJ protein families can be distinguished, claudins, comprising 27 members in mammals, and TJ-associated MARVEL proteins (TAMP), comprising occludin, tricellulin, and MarvelD3. They are linked to a multitude of TJ-associated regulatory and scaffolding proteins. The major TJ proteins are classified according to the physiological role they play in enabling or preventing paracellular transport. Many TJ proteins have sealing functions (claudins 1, 3, 5, 11, 14, 19, and tricellulin). In contrast, a significant number of claudins form channels across TJs which feature selectivity for cations (claudins 2, 10b, and 15), anions (claudin-10a and -17), or are permeable to water (claudin-2). For several TJ proteins, function is yet unclear as their effects on epithelial barriers are inconsistent (claudins 4, 7, 8, 16, and occludin). TJs undergo physiological and pathophysiological regulation by altering protein composition or abundance. Major pathophysiological conditions which involve changes in TJ protein composition are (1) effects of pathogens binding to TJ proteins, (2) altered TJ protein composition during inflammation and infection, and (3) altered TJ protein expression in cancers.
Collapse
Affiliation(s)
- Dorothee Günzel
- Institute of Clinical Physiology, Charité, Universtätsmedizin Berlin, Campus Benjamin Franklin, Freie Universität/Humboldt-Universität, Berlin, Germany
| | | |
Collapse
|
28
|
Schindler RF, Poon KL, Simrick S, Brand T. The Popeye domain containing genes: essential elements in heart rate control. Cardiovasc Diagn Ther 2013; 2:308-19. [PMID: 24282731 DOI: 10.3978/j.issn.2223-3652.2012.12.01] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 12/06/2012] [Indexed: 01/25/2023]
Abstract
The Popeye domain containing (Popdc) gene family displays preferential expression in skeletal muscle and heart. Only recently a significant gain in the understanding of the function of Popdc genes in the heart has been obtained. The Popdc genes encode membrane proteins harboring an evolutionary conserved Popeye domain, which functions as a binding domain for cyclic adenosine monophosphate (cAMP). Popdc proteins interact with the two-pore channel TREK-1 and enhance its current. This protein interaction is modulated by cAMP. Null mutations of members of the Popdc gene family in zebrafish and mouse are associated with severe cardiac arrhythmia phenotypes. While in zebrafish an atrioventricular block was prevalent, in mouse a stress-induced sinus bradycardia was observed, which was due to the presence of sinus pauses. Moreover, the phenotype develops in an age-dependent manner, being absent in the young animal and becoming increasingly severe, as the animals grow older. This phenotype is reminiscent of the sick sinus syndrome (SSS), which affects mostly the elderly and is characterized by the poor ability of the cardiac pacemaker to adapt the heart rate to the physiological demand. While being a prevalent disease, which is responsible for a large fraction of pacemaker implantations in Western countries, SSS is poorly understood at the molecular level. It is therefore expected that the study of the molecular basis of the stress-induced bradycardia in Popdc mice will shed new light on the etiology of pacemaker disease.
Collapse
Affiliation(s)
- Roland F Schindler
- Heart Science Centre, Imperial College London, Harefield, United Kingdom
| | | | | | | |
Collapse
|
29
|
Benesh EC, Miller PM, Pfaltzgraff ER, Grega-Larson NE, Hager HA, Sung BH, Qu X, Baldwin HS, Weaver AM, Bader DM. Bves and NDRG4 regulate directional epicardial cell migration through autocrine extracellular matrix deposition. Mol Biol Cell 2013; 24:3496-510. [PMID: 24048452 PMCID: PMC3826988 DOI: 10.1091/mbc.e12-07-0539] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The Bves and NDRG4 proteins interact to regulate directional cell movement by mediating cell surface fusion of internalized fibronectin for resecretion. This provides the first evidence of Bves/NDRG4 protein function within subcellular trafficking pathways and explains how the Bves complex diversely influences development, cancer, and repair. Directional cell movement is universally required for tissue morphogenesis. Although it is known that cell/matrix interactions are essential for directional movement in heart development, the mechanisms governing these interactions require elucidation. Here we demonstrate that a novel protein/protein interaction between blood vessel epicardial substance (Bves) and N-myc downstream regulated gene 4 (NDRG4) is critical for regulation of epicardial cell directional movement, as disruption of this interaction randomizes migratory patterns. Our studies show that Bves/NDRG4 interaction is required for trafficking of internalized fibronectin through the “autocrine extracellular matrix (ECM) deposition” fibronectin recycling pathway. Of importance, we demonstrate that Bves/NDRG4-mediated fibronectin recycling is indeed essential for epicardial cell directional movement, thus linking these two cell processes. Finally, total internal reflectance fluorescence microscopy shows that Bves/NDRG4 interaction is required for fusion of recycling endosomes with the basal cell surface, providing a molecular mechanism of motility substrate delivery that regulates cell directional movement. This is the first evidence of a molecular function for Bves and NDRG4 proteins within broader subcellular trafficking paradigms. These data identify novel regulators of a critical vesicle-docking step required for autocrine ECM deposition and explain how Bves facilitates cell-microenvironment interactions in the regulation of epicardial cell–directed movement.
Collapse
Affiliation(s)
- Emily C Benesh
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Pediatric Cardiology, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Pathology, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232 Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO 63110
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Simrick S, Schindler RF, Poon KL, Brand T. Popeye domain-containing proteins and stress-mediated modulation of cardiac pacemaking. Trends Cardiovasc Med 2013; 23:257-63. [PMID: 23562093 DOI: 10.1016/j.tcm.2013.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 10/27/2022]
Abstract
An intricate network of ion channels and pumps are involved in generating a diastolic pacemaker potential, which is transmitted to the working myocardium with the help of the cardiac conduction system. The principles of cardiac pacemaking are reasonably well understood, however, the mechanism by which the heart increases its beating frequency in response to adrenergic stimulation has not been fully worked out. The Popeye domain-containing (Popdc) genes encode plasma membrane-localized proteins that are able to bind cAMP with high affinity; mice with null mutations in Popdc1 or 2 have a stress-induced pacemaker dysfunction. The phenotype in both mutants develops in an age-dependent manner and thus may model pacemaker dysfunction in man, as well as provide novel mechanistic insights into the process of pacemaker adaptation to stress.
Collapse
Affiliation(s)
- Subreena Simrick
- Heart Science Centre, National Heart and Lung Institute, Imperial College London, Harefield UB9 6JH, United Kingdom
| | | | | | | |
Collapse
|
31
|
Wu M, Li Y, He X, Shao X, Yang F, Zhao M, Wu C, Zhang C, Zhou L. Mutational and functional analysis of the BVES gene coding region in Chinese patients with non-syndromic tetralogy of Fallot. Int J Mol Med 2013; 31:899-903. [PMID: 23403794 DOI: 10.3892/ijmm.2013.1275] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 12/21/2012] [Indexed: 11/06/2022] Open
Abstract
Tetralogy of Fallot (TOF) is a common congenital heart defect (CHD). However, the genetic causes are largely unknown. Blood vessel epicardial substance (BVES) is postulated to play a role in embryonic development, and we previously found that the expression of BVES is upregulation in patients with congenital septal defect and that BVES participates in cardiocyte differentiation. We hypothesized that BVES is a candidate gene of TOF. In the present study, the entire coding sequence and splice junctions of BVES were sequenced in 114 unrelated patients with TOF and 400 unrelated healthy individuals used as controls. The functional effects of the exon mutant BVES were characterized in contrast to its wild-type counterpart using a luciferase reporter assay system. Four novel BVES mutations (c.166T>C, c.909C>T, c.540-80C>T, c.958+30A>G) were identified in patients with TOF but not in the 400 controls. Functional analysis indicated that the c.166T>C mutation of BVES was associated with an increased transcriptional activity. This finding suggests that BVES as a novel TOF gene may provide further insight into the molecular mechanisms involved in TOF.
Collapse
Affiliation(s)
- Min Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Schulzke JD, Günzel D, John LJ, Fromm M. Perspectives on tight junction research. Ann N Y Acad Sci 2012; 1257:1-19. [PMID: 22671584 DOI: 10.1111/j.1749-6632.2012.06485.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The tight junction connects neighboring epithelial or endothelial cells. As a general function, it seals the paracellular pathway and thus prevents back-leakage of just transported solutes and water. However, not all tight junctions are merely tight: some tight junction proteins build their own transport pathways by forming channels selective for small cations, anions, or water. Two families of tight junction proteins have been identified, claudins (27 members in mammals) and tight junction-associated MARVEL proteins ((TAMPs) occludin, tricellulin, and MarvelD3); an additional, structurally different, junction protein is junction adhesion molecule (JAM). Besides classification by genetic or molecular kinship, classification of tight junction proteins has been suggested according to permeability attributes. Recent studies describe specific cis and trans interactions and manifold physiologic regulations of claudins and TAMPs. In many inflammatory and infectious diseases they are found to be altered, for example, causing adversely increased permeability. Currently, attempts are being made to alter the paracellular barrier for therapeutic interventions or for transiently facilitating drug uptake. This overview concludes with a list of open questions and future topics in tight junction research.
Collapse
Affiliation(s)
- Jörg-Dieter Schulzke
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Division of Nutritional Medicine, Charité, Universitätsmedizin Berlin, Germany.
| | | | | | | |
Collapse
|
33
|
Abstract
The epithelial tight junction (TJ) is the apical-most intercellular junction and serves as a gatekeeper for the paracellular pathway by permitting regulated passage of fluid and ions while restricting movement of large molecules. In addition to these vital barrier functions, TJ proteins are emerging as major signaling molecules that mediate crosstalk between the extracellular environment, the cell surface, and the nucleus. Biochemical studies have recently determined that epithelial TJs contain over a hundred proteins that encompass transmembrane proteins, scaffolding molecules, cytoskeletal components, regulatory elements, and signaling molecules. Indeed, many of these proteins have defined roles in regulating epithelial polarity, differentiation, and proliferation. This review will focus on recent findings that highlight a role for TJ proteins in controlling cell proliferation during epithelial homeostasis, wound healing, and carcinogenesis.
Collapse
Affiliation(s)
- Attila E Farkas
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
34
|
Expression and distribution of symplekin regulates the assembly and function of the epithelial tight junction. Histochem Cell Biol 2012; 137:319-27. [DOI: 10.1007/s00418-011-0906-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2011] [Indexed: 12/19/2022]
|
35
|
Luo D, Huang H, Lu ML, Zhao GF, Chang J, Zheng MY, Wang Y. Abnormal Expression of Adhesion Protein Bves is Associated with Gastric Cancer Progression and Poor Survival. Pathol Oncol Res 2011; 18:491-7. [DOI: 10.1007/s12253-011-9472-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Accepted: 10/27/2011] [Indexed: 12/20/2022]
|
36
|
Possible involvement of tight junctions, extracellular matrix and nuclear receptors in epithelial differentiation. J Biomed Biotechnol 2011; 2011:253048. [PMID: 22162632 PMCID: PMC3227411 DOI: 10.1155/2011/253048] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 08/28/2011] [Accepted: 09/09/2011] [Indexed: 01/16/2023] Open
Abstract
Tight junctions are intercellular junctions localized at the most apical end of the lateral plasma membrane. They consist of four kinds of transmembrane proteins (occludin, claudins, junctional adhesion molecules, and tricellulin) and huge numbers of scaffolding proteins and contribute to the paracellular barrier and fence function. The mutation and deletion of these proteins impair the functions of tight junctions and cause various human diseases. In this paper, we provide an overview of recent studies on transmembrane proteins of tight junctions and highlight the functional significance of tight junctions, extracellular matrix, and nuclear receptors in epithelial differentiation.
Collapse
|