Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Publication year range
1.
EMBO J ; 43(14): 2878-2907, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38816652

ABSTRACT

In mice, γδ-T lymphocytes that express the co-stimulatory molecule, CD27, are committed to the IFNγ-producing lineage during thymic development. In the periphery, these cells play a critical role in host defense and anti-tumor immunity. Unlike αß-T cells that rely on MHC-presented peptides to drive their terminal differentiation, it is unclear whether MHC-unrestricted γδ-T cells undergo further functional maturation after exiting the thymus. Here, we provide evidence of phenotypic and functional diversity within peripheral IFNγ-producing γδ T cells. We found that CD27+ Ly6C- cells convert into CD27+Ly6C+ cells, and these CD27+Ly6C+ cells control cancer progression in mice, while the CD27+Ly6C- cells cannot. The gene signatures of these two subsets were highly analogous to human immature and mature γδ-T cells, indicative of conservation across species. We show that IL-27 supports the cytotoxic phenotype and function of mouse CD27+Ly6C+ cells and human Vδ2+ cells, while IL-27 is dispensable for mouse CD27+Ly6C- cell and human Vδ1+ cell functions. These data reveal increased complexity within IFNγ-producing γδ-T cells, comprising immature and terminally differentiated subsets, that offer new insights into unconventional T-cell biology.


Subject(s)
Antigens, Ly , Receptors, Antigen, T-Cell, gamma-delta , Tumor Necrosis Factor Receptor Superfamily, Member 7 , Animals , Mice , Antigens, Ly/metabolism , Antigens, Ly/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Humans , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Interferon-gamma/metabolism , Interferon-gamma/immunology , Interleukin-27/metabolism , Interleukin-27/genetics , Cell Differentiation/immunology , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
2.
EMBO J ; 42(18): e113987, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37577760

ABSTRACT

Dysregulation of the PI3K/AKT pathway is a common occurrence in high-grade serous ovarian carcinoma (HGSOC), with the loss of the tumour suppressor PTEN in HGSOC being associated with poor prognosis. The cellular mechanisms of how PTEN loss contributes to HGSOC are largely unknown. We here utilise time-lapse imaging of HGSOC spheroids coupled to a machine learning approach to classify the phenotype of PTEN loss. PTEN deficiency induces PI(3,4,5)P3 -rich and -dependent membrane protrusions into the extracellular matrix (ECM), resulting in a collective invasion phenotype. We identify the small GTPase ARF6 as a crucial vulnerability of HGSOC cells upon PTEN loss. Through a functional proteomic CRISPR screen of ARF6 interactors, we identify the ARF GTPase-activating protein (GAP) AGAP1 and the ECM receptor ß1-integrin (ITGB1) as key ARF6 interactors in HGSOC regulating PTEN loss-associated invasion. ARF6 functions to promote invasion by controlling the recycling of internalised, active ß1-integrin to maintain invasive activity into the ECM. The expression of the CYTH2-ARF6-AGAP1 complex in HGSOC patients is inversely associated with outcome, allowing the identification of patient groups with improved versus poor outcome. ARF6 may represent a therapeutic vulnerability in PTEN-depleted HGSOC.


Subject(s)
Monomeric GTP-Binding Proteins , Ovarian Neoplasms , Humans , Female , Integrins/metabolism , Proteomics , Phosphatidylinositol 3-Kinases/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Monomeric GTP-Binding Proteins/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
3.
J Mol Cell Cardiol ; 186: 71-80, 2024 01.
Article in English | MEDLINE | ID: mdl-37956903

ABSTRACT

Gap junction and ion channel remodeling occur early in Arrhythmogenic Cardiomyopathy (ACM), but their pathogenic consequences have not been elucidated. Here, we identified the arrhythmogenic substrate, consisting of propagation slowing and conduction block, in ACM models expressing two different desmosomal gene variants. Neonatal rat ventricular myocytes were transduced to express variants in genes encoding desmosomal proteins plakoglobin or plakophilin-2. Studies were performed in engineered cells and anisotropic tissues to quantify changes in conduction velocity, formation of unidirectional propagation, cell-cell electrical coupling, and ion currents. Conduction velocity decreased by 71% and 63% in the two ACM models. SB216763, an inhibitor of glycogen synthase kinase-3 beta, restored conduction velocity to near normal levels. Compared to control, both ACM models showed greater propensity for unidirectional conduction block, which increased further at greater stimulation frequencies. Cell-cell electrical conductance measured in cell pairs was reduced by 86% and 87% in the two ACM models. Computer modeling showed close correspondence between simulated and experimentally determined changes in conduction velocity. The simulation identified that reduced cell-cell electrical coupling was the dominant factor leading to slow conduction, while the combination of reduced cell-cell electrical coupling, reduced sodium current and inward rectifier potassium current explained the development of unidirectional block. Expression of two different ACM variants markedly reduced cell-cell electrical coupling and conduction velocity, and greatly increased the likelihood of developing unidirectional block - both key features of arrhythmogenesis. This study provides the first quantitative analysis of cellular electrophysiological changes leading to the substrate of reentrant arrhythmias in early stage ACM.


Subject(s)
Cardiomyopathies , Myocytes, Cardiac , Rats , Animals , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/metabolism , Gap Junctions/metabolism , Ion Channels/metabolism , Cardiomyopathies/metabolism
4.
Physiol Rev ; 97(1): 227-252, 2017 01.
Article in English | MEDLINE | ID: mdl-27881552

ABSTRACT

Unique to striated muscle cells, transverse tubules (t-tubules) are membrane organelles that consist of sarcolemma penetrating into the myocyte interior, forming a highly branched and interconnected network. Mature t-tubule networks are found in mammalian ventricular cardiomyocytes, with the transverse components of t-tubules occurring near sarcomeric z-discs. Cardiac t-tubules contain membrane microdomains enriched with ion channels and signaling molecules. The microdomains serve as key signaling hubs in regulation of cardiomyocyte function. Dyad microdomains formed at the junctional contact between t-tubule membrane and neighboring sarcoplasmic reticulum are critical in calcium signaling and excitation-contraction coupling necessary for beat-to-beat heart contraction. In this review, we provide an overview of the current knowledge in gross morphology and structure, membrane and protein composition, and function of the cardiac t-tubule network. We also review in detail current knowledge on the formation of functional membrane subdomains within t-tubules, with a particular focus on the cardiac dyad microdomain. Lastly, we discuss the dynamic nature of t-tubules including membrane turnover, trafficking of transmembrane proteins, and the life cycles of membrane subdomains such as the cardiac BIN1-microdomain, as well as t-tubule remodeling and alteration in diseased hearts. Understanding cardiac t-tubule biology in normal and failing hearts is providing novel diagnostic and therapeutic opportunities to better treat patients with failing hearts.


Subject(s)
Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Animals , Humans
5.
Br J Cancer ; 128(11): 1991-1999, 2023 06.
Article in English | MEDLINE | ID: mdl-36991255

ABSTRACT

BACKGROUND: Prostate cancer is the most common cancer in men in the developed world, with most deaths caused by advanced and metastatic disease which has no curative options. Here, we identified Mbtps2 alteration to be associated with metastatic disease in an unbiased in vivo screen and demonstrated its regulation of fatty acid and cholesterol metabolism. METHODS: The Sleeping Beauty transposon system was used to randomly alter gene expression in the PtenNull murine prostate. MBTPS2 was knocked down by siRNA in LNCaP, DU145 and PC3 cell lines, which were then phenotypically investigated. RNA-Seq was performed on LNCaP cells lacking MBTPS2, and pathways validated by qPCR. Cholesterol metabolism was investigated by Filipin III staining. RESULTS: Mbtps2 was identified in our transposon-mediated in vivo screen to be associated with metastatic prostate cancer. Silencing of MBTPS2 expression in LNCaP, DU145 and PC3 human prostate cancer cells reduced proliferation and colony forming growth in vitro. Knockdown of MBTPS2 expression in LNCaP cells impaired cholesterol synthesis and uptake along with reduced expression of key regulators of fatty acid synthesis, namely FASN and ACACA. CONCLUSION: MBTPS2 is implicated in progressive prostate cancer and may mechanistically involve its effects on fatty acid and cholesterol metabolism.


Subject(s)
Lipogenesis , Prostatic Neoplasms , Male , Humans , Animals , Mice , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Cell Line, Tumor , Prostatic Neoplasms/pathology , Cholesterol , Fatty Acids , Cell Proliferation , Gene Expression Regulation, Neoplastic , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism
6.
J Hepatol ; 78(5): 1028-1036, 2023 05.
Article in English | MEDLINE | ID: mdl-36702176

ABSTRACT

BACKGROUND & AIMS: Mouse models of lineage tracing have helped to describe the important subpopulations of hepatocytes responsible for liver regeneration. However, conflicting results have been obtained from different models. Herein, we aimed to reconcile these conflicting reports by repeating a key lineage-tracing study from pericentral hepatocytes and characterising this Axin2CreERT2 model in detail. METHODS: We performed detailed characterisation of the labelled population in the Axin2CreERT2 model. We lineage traced this cell population, quantifying the labelled population over 1 year and performed in-depth phenotypic comparisons, including transcriptomics, metabolomics and analysis of proteins through immunohistochemistry, of Axin2CreERT2 mice to WT counterparts. RESULTS: We found that after careful definition of a baseline population, there are marked differences in labelling between male and female mice. Upon induced lineage tracing there was no expansion of the labelled hepatocyte population in Axin2CreERT2 mice. We found substantial evidence of disrupted homeostasis in Axin2CreERT2 mice. Offspring are born with sub-Mendelian ratios and adult mice have perturbations of hepatic Wnt/ß-catenin signalling and related metabolomic disturbance. CONCLUSIONS: We find no evidence of predominant expansion of the pericentral hepatocyte population during liver homeostatic regeneration. Our data highlight the importance of detailed preclinical model characterisation and the pitfalls which may occur when comparing across sexes and backgrounds of mice and the effects of genetic insertion into native loci. IMPACT AND IMPLICATIONS: Understanding the source of cells which regenerate the liver is crucial to harness their potential to regrow injured livers. Herein, we show that cells which were previously thought to repopulate the liver play only a limited role in physiological regeneration. Our data helps to reconcile differing conclusions drawn from results from a number of prior studies and highlights methodological challenges which are relevant to preclinical models more generally.


Subject(s)
Focal Nodular Hyperplasia , Liver Regeneration , Male , Female , Humans , Liver Regeneration/physiology , Hepatocytes/metabolism , Liver/metabolism , Homeostasis , Cell Proliferation , Axin Protein/genetics
7.
Am J Physiol Heart Circ Physiol ; 324(6): H751-H761, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36961487

ABSTRACT

Preclinical large animal models of chronic heart failure (HF) are crucial to both understanding pathological remodeling and translating fundamental discoveries into novel therapeutics for HF. Canine models of ischemic cardiomyopathy are historically limited by either high early mortality or failure to develop chronic heart failure. Twenty-nine healthy adult dogs (30 ± 4 kg, 15/29 male) underwent thoracotomy followed by one of three types of left anterior descending (LAD) coronary artery ligation procedures: group 1 (n = 4) (simple LAD: proximal and distal LAD ligation); group 2 (n = 14) (simple LAD plus lateral wall including ligation of the distal first diagonal and proximal first obtuse marginal); and group 3 (n = 11) (total LAD devascularization or TLD: simple LAD plus ligation of proximal LAD branches to both the right and left ventricles). Dogs were followed until chronic severe HF developed defined as left ventricular ejection fraction (LVEF) < 40% and NH2-terminal-prohormone B-type natriuretic peptide (NT-proBNP) > 900 pmol/L. Overall early survival (48-h postligation) in 29 dogs was 83% and the survival rate at postligation 5 wk was 69%. Groups 1 and 2 had 100% and 71% early survival, respectively, yet only a 50% success rate of developing chronic HF. Group 3 had excellent survival at postligation 48 h (91%) and a 100% success in the development of chronic ischemic HF. The TLD approach, which limits full LAD and collateral flow to its perfusion bed, provides excellent early survival and reliable development of chronic ischemic HF in canine hearts.NEW & NOTEWORTHY The novel total left anterior descending devascularization (TLD) approach in a canine ischemic heart failure model limits collateral flow in the ischemic zone and provides excellent early survival and repeatable development of chronic ischemic heart failure in the canine heart. This work provides a consistent large animal model for investigating heart failure mechanisms and testing novel therapeutics.


Subject(s)
Heart Failure , Ventricular Function, Left , Dogs , Male , Animals , Stroke Volume , Heart Failure/etiology , Heart , Chronic Disease , Disease Models, Animal
8.
BMC Bioinformatics ; 22(1): 411, 2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34412594

ABSTRACT

BACKGROUND: Once bulk RNA-seq data has been processed, i.e. aligned and then expression and differential tables generated, there remains the essential process where the biology is explored, visualized and interpreted. Without the use of a visualisation and interpretation pipeline this step can be time consuming and laborious, and is often completed using R. Though commercial visualisation and interpretation pipelines are comprehensive, freely available pipelines are currently more limited. RESULTS: Here we demonstrate Searchlight, a freely available bulk RNA-seq visualisation and interpretation pipeline. Searchlight provides: a comprehensive statistical and visual analysis, focusing on the global, pathway and single gene levels; compatibility with most differential experimental designs irrespective of organism or experimental complexity, via three workflows; reports; and support for downstream user modification of plots via user-friendly R-scripts and a Shiny app. We show that Searchlight offers greater automation than current best tools (VIPER and BioJupies). We demonstrate in a timed re-analysis study, that alongside a standard bulk RNA-seq processing pipeline, Searchlight can be used to complete bulk RNA-seq projects up to the point of manuscript quality figures, in under 3 h. CONCLUSIONS: Compared to a manual R based analysis or current best freely available pipelines (VIPER and BioJupies), Searchlight can reduce the time and effort needed to complete bulk RNA-seq projects to manuscript level. Searchlight is suitable for bioinformaticians, service providers and bench scientists. https://github.com/Searchlight2/Searchlight2 .


Subject(s)
Publications , Software , RNA-Seq , Exome Sequencing , Workflow
9.
PLoS Biol ; 15(8): e2002354, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28806752

ABSTRACT

Microparticles (MPs) are cell-cell communication vesicles derived from the cell surface plasma membrane, although they are not known to originate from cardiac ventricular muscle. In ventricular cardiomyocytes, the membrane deformation protein cardiac bridging integrator 1 (cBIN1 or BIN1+13+17) creates transverse-tubule (t-tubule) membrane microfolds, which facilitate ion channel trafficking and modulate local ionic concentrations. The microfold-generated microdomains continuously reorganize, adapting in response to stress to modulate the calcium signaling apparatus. We explored the possibility that cBIN1-microfolds are externally released from cardiomyocytes. Using electron microscopy imaging with immunogold labeling, we found in mouse plasma that cBIN1 exists in membrane vesicles about 200 nm in size, which is consistent with the size of MPs. In mice with cardiac-specific heterozygous Bin1 deletion, flow cytometry identified 47% less cBIN1-MPs in plasma, supporting cardiac origin. Cardiac release was also evidenced by the detection of cBIN1-MPs in medium bathing a pure population of isolated adult mouse cardiomyocytes. In human plasma, osmotic shock increased cBIN1 detection by enzyme-linked immunosorbent assay (ELISA), and cBIN1 level decreased in humans with heart failure, a condition with reduced cardiac muscle cBIN1, both of which support cBIN1 release in MPs from human hearts. Exploring putative mechanisms of MP release, we found that the membrane fission complex endosomal sorting complexes required for transport (ESCRT)-III subunit charged multivesicular body protein 4B (CHMP4B) colocalizes and coimmunoprecipitates with cBIN1, an interaction enhanced by actin stabilization. In HeLa cells with cBIN1 overexpression, knockdown of CHMP4B reduced the release of cBIN1-MPs. Using truncation mutants, we identified that the N-terminal BAR (N-BAR) domain in cBIN1 is required for CHMP4B binding and MP release. This study links the BAR protein superfamily to the ESCRT pathway for MP biogenesis in mammalian cardiac ventricular cells, identifying elements of a pathway by which cytoplasmic cBIN1 is released into blood.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell-Derived Microparticles/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/blood , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/blood , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/ultrastructure , Enzyme-Linked Immunosorbent Assay , Exons , HeLa Cells , Heart Failure/blood , Heart Failure/pathology , Heterozygote , Humans , Mice, Transgenic , Microscopy, Electron, Transmission , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Proteins/blood , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Particle Size , Peptide Fragments/blood , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/blood , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
11.
Circ Res ; 121(9): 1069-1080, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-28923791

ABSTRACT

RATIONALE: Delivery of Cx43 (connexin 43) to the intercalated disc is a continuous and rapid process critical for intercellular coupling. By a pathway of targeted delivery involving microtubule highways, vesicles of Cx43 hemichannels are efficiently trafficked to adherens junctions at intercalated discs. It has also been identified that actin provides rest stops for Cx43 forward trafficking and that Cx43 has a 20 kDa internally translated small C terminus isoform, GJA1-20k (Gap Junction Protein Alpha 1- 20 kDa), which is required for full-length Cx43 trafficking, but by an unknown mechanism. OBJECTIVE: We explored the mechanism by which the GJA1-20k isoform is required for full-length Cx43 forward trafficking to intercalated discs. METHODS AND RESULTS: Using an in vivo Adeno-associated virus serotype 9-mediated gene transfer system, we confirmed in whole animal that GJA1-20k markedly increases endogenous myocardial Cx43 gap junction plaque size at the intercalated discs. In micropatterned cell pairing systems, we found that exogenous GJA1-20k expression stabilizes filamentous actin without affecting actin protein expression and that GJA1-20k complexes with both actin and tubulin. We also found that filamentous actin regulates microtubule organization as inhibition of actin polymerization with a low dose of latrunculin A disrupts the targeting of microtubules to cell-cell junctions. GJA1-20k protects actin filament from latrunculin A disruption, preserving microtubule trajectory to the cell-cell border. For therapeutic implications, we found that prior in vivo Adeno-associated virus serotype 9-mediated gene delivery of GJA1-20k to the heart protects Cx43 localization to the intercalated discs against acute ischemic injury. CONCLUSIONS: The internally translated GJA1-20k isoform stabilizes actin filaments, which guides growth trajectories of the Cx43 microtubule trafficking machinery, increasing delivery of Cx43 hemichannels to cardiac intercalated discs. Exogenous GJA1-20k helps to maintain cell-cell coupling in instances of anticipated myocardial ischemia.


Subject(s)
Actins/metabolism , Connexin 43/metabolism , Gene Transfer Techniques , Myocytes, Cardiac/metabolism , Actins/genetics , Animals , Animals, Newborn , Cells, Cultured , Connexin 43/genetics , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Microtubules/genetics , Microtubules/metabolism , Organ Culture Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/physiology
12.
Biochim Biophys Acta Biomembr ; 1860(1): 40-47, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28576298

ABSTRACT

Gap Junction (GJ) channels, including the most common Connexin 43 (Cx43), have fundamental roles in excitable tissues by facilitating rapid transmission of action potentials between adjacent cells. For instance, synchronization during each heartbeat is regulated by these ion channels at the cardiomyocyte cell-cell border. Cx43 protein has a short half-life, and rapid synthesis and timely delivery of those proteins to particular subdomains are crucial for the cellular organization of gap junctions and maintenance of intracellular coupling. Impairment in gap junction trafficking contributes to dangerous complications in diseased hearts such as the arrhythmias of sudden cardiac death. Of recent interest are the protein-protein interactions with the Cx43 carboxy-terminus. These interactions have significant impact on the full length Cx43 lifecycle and also contribute to trafficking of Cx43 as well as possibly other functions. We are learning that many of the known non-canonical roles of Cx43 can be attributed to the recently identified six endogenous Cx43 truncated isoforms which are produced by internal translation. In general, alternative translation is a new leading edge for proteome expansion and therapeutic drug development. This review highlights recent mechanisms identified in the trafficking of gap junction channels, involvement of other proteins contributing to the delivery of channels to the cell-cell border, and understanding of possible roles of the newly discovered alternatively translated isoforms in Cx43 biology. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.


Subject(s)
Arrhythmias, Cardiac/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Ion Channels/metabolism , Myocytes, Cardiac/metabolism , Protein Biosynthesis , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Connexin 43/genetics , Death, Sudden, Cardiac , Gap Junctions/genetics , Gap Junctions/pathology , Humans , Ion Channels/genetics , Myocytes, Cardiac/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport
13.
Biochim Biophys Acta ; 1863(7 Pt B): 1848-56, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26526689

ABSTRACT

With each heartbeat, Connexin43 (Cx43) cell-cell communication gap junctions are needed to rapidly spread and coordinate excitation signals for an effective heart contraction. The correct formation and delivery of channels to their respective membrane subdomain is referred to as protein trafficking. Altered Cx43 trafficking is a dangerous complication of diseased myocardium which contributes to the arrhythmias of sudden cardiac death. Cx43 has also been found to regulate many other cellular processes that cannot be explained by cell-cell communication. We recently identified the existence of up to six endogenous internally translated Cx43 N-terminal truncated isoforms from the same full-length mRNA molecule. This is the first evidence that alternative translation is possible for human ion channels and in human heart. Interestingly, we found that these internally translated isoforms, more specifically the 20 kDa isoform (GJA1-20k), is important for delivery of Cx43 to its respective membrane subdomain. This review covers recent advances in Cx43 trafficking and potential importance of alternatively translated Cx43 truncated isoforms. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Connexin 43/metabolism , Gap Junctions/metabolism , Membrane Microdomains/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Animals , Cell Communication , Connexin 43/chemistry , Connexin 43/genetics , Endocytosis , Genetic Predisposition to Disease , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/physiopathology , Humans , Myocardial Contraction , Protein Conformation , Protein Isoforms , Protein Transport , Structure-Activity Relationship
14.
Traffic ; 15(6): 684-99, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24612377

ABSTRACT

Altered phosphorylation and trafficking of connexin 43 (Cx43) during acute ischemia contributes to arrhythmogenic gap junction remodeling, yet the critical sequence and accessory proteins necessary for Cx43 internalization remain unresolved. 14-3-3 proteins can regulate protein trafficking, and a 14-3-3 mode-1 binding motif is activated upon phosphorylation of Ser373 of the Cx43 C-terminus. We hypothesized that Cx43(Ser373) phosphorylation is important to pathological gap junction remodeling. Immunofluorescence in human heart reveals the enrichment of 14-3-3 proteins at intercalated discs, suggesting interaction with gap junctions. Knockdown of 14-3-3τ in cell lines increases gap junction plaque size at cell-cell borders. Cx43(S373A) mutation prevents Cx43/14-3-3 complexing and stabilizes Cx43 at the cell surface, indicating avoidance of degradation. Using Langendorff-perfused mouse hearts, we detect phosphorylation of newly internalized Cx43 at Ser373 and Ser368 within 30 min of no-flow ischemia. Phosphorylation of Cx43 at Ser368 by protein kinase C and Ser255 by mitogen-activated protein kinase has previously been implicated in Cx43 internalization. The Cx43(S373A) mutant is resistant to phosphorylation at both these residues and does not undergo ubiquitination, revealing Ser373 phosphorylation as an upstream gatekeeper of a posttranslational modification cascade necessary for Cx43 internalization. Cx43(Ser373) phosphorylation is a potent target for therapeutic interventions to preserve gap junction coupling in the stressed myocardium.


Subject(s)
14-3-3 Proteins/metabolism , Connexin 43/metabolism , Endocytosis , Gap Junctions/metabolism , Myocardial Reperfusion Injury/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , Amino Acid Motifs , Animals , Binding Sites , Cell Line , Connexin 43/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Protein Stability , Protein Transport
15.
PLoS Biol ; 11(12): e1001727, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24311987

ABSTRACT

The 3-O-sulfotransferase (3-OST) family catalyzes rare modifications of glycosaminoglycan chains on heparan sulfate proteoglycans, yet their biological functions are largely unknown. Knockdown of 3-OST-7 in zebrafish uncouples cardiac ventricular contraction from normal calcium cycling and electrophysiology by reducing tropomyosin4 (tpm4) expression. Normal 3-OST-7 activity prevents the expansion of BMP signaling into ventricular myocytes, and ectopic activation of BMP mimics the ventricular noncontraction phenotype seen in 3-OST-7 depleted embryos. In 3-OST-7 morphants, ventricular contraction can be rescued by overexpression of tropomyosin tpm4 but not by troponin tnnt2, indicating that tpm4 serves as a lynchpin for ventricular sarcomere organization downstream of 3-OST-7. Contraction can be rescued by expression of 3-OST-7 in endocardium, or by genetic loss of bmp4. Strikingly, BMP misregulation seen in 3-OST-7 morphants also occurs in multiple cardiac noncontraction models, including potassium voltage-gated channel gene, kcnh2, affected in Romano-Ward syndrome and long-QT syndrome, and cardiac troponin T gene, tnnt2, affected in human cardiomyopathies. Together these results reveal 3-OST-7 as a key component of a novel pathway that constrains BMP signaling from ventricular myocytes, coordinates sarcomere assembly, and promotes cardiac contractile function.


Subject(s)
Bone Morphogenetic Proteins/physiology , Myocardial Contraction/physiology , Sulfotransferases/physiology , Zebrafish Proteins/physiology , Action Potentials/physiology , Animals , Gene Knockdown Techniques , Muscle Development/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Signal Transduction/physiology , Tropomyosin/physiology , Zebrafish
17.
Circ Res ; 114(6): 982-92, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24508725

ABSTRACT

RATIONALE: Kv1.5 (KCNA5) mediates the ultra-rapid delayed rectifier current that controls atrial action potential duration. Given its atrial-specific expression and alterations in human atrial fibrillation, Kv1.5 has emerged as a promising target for the treatment of atrial fibrillation. A necessary step in the development of novel agents that selectively modulate trafficking pathways is the identification of the cellular machinery controlling Kv1.5 surface density, of which little is yet known. OBJECTIVE: To investigate the role of the unconventional myosin-V (MYO5A and MYO5B) motors in determining the cell surface density of Kv1.5. METHODS AND RESULTS: Western blot analysis showed MYO5A and MYO5B expression in the heart, whereas disruption of endogenous motors selectively reduced IKur current in adult rat cardiomyocytes. Dominant negative constructs and short hairpin RNA silencing demonstrated a role for MYO5A and MYO5B in the surface trafficking of Kv1.5 and connexin-43 but not potassium voltage-gated channel, subfamily H (eag-related), member 2 (KCNH2). Live-cell imaging of Kv1.5-GFP and retrospective labeling of phalloidin demonstrated motility of Kv1.5 vesicles on actin tracts. MYO5A participated in anterograde trafficking, whereas MYO5B regulated postendocytic recycling. Overexpression of mutant motors revealed a selective role for Rab11 in coupling MYO5B to Kv1.5 recycling. CONCLUSIONS: MYO5A and MYO5B control functionally distinct steps in the surface trafficking of Kv1.5. These isoform-specific trafficking pathways determine Kv1.5-encoded IKur in myocytes to regulate repolarizing current and, consequently, cardiac excitability. Therapeutic strategies that manipulate Kv1.5 selective trafficking pathways may prove useful in the treatment of arrhythmias.


Subject(s)
Cell Membrane/metabolism , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/physiology , Myosin Type V/physiology , Myosins/physiology , Protein Transport/physiology , Actin Cytoskeleton/physiology , Animals , Arrhythmias, Cardiac/physiopathology , Cell Line , Connexin 43/analysis , ERG1 Potassium Channel , Endocytosis , Ether-A-Go-Go Potassium Channels/analysis , Gap Junctions , Genes, Reporter , Heart Conduction System/physiopathology , Ion Transport , Kv1.5 Potassium Channel/genetics , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Models, Cardiovascular , Myosin Heavy Chains/deficiency , Myosin Heavy Chains/genetics , Myosin Type V/deficiency , Myosin Type V/genetics , Myosins/deficiency , Myosins/genetics , Potassium/metabolism , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , rab GTP-Binding Proteins/physiology
18.
J Physiol ; 593(6): 1347-60, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25772290

ABSTRACT

This paper is the second of a series of three reviews published in this issue resulting from the University of California Davis Cardiovascular Symposium 2014: Systems approach to understanding cardiac excitation-contraction coupling and arrhythmias: Na(+) channel and Na(+) transport. The goal of the symposium was to bring together experts in the field to discuss points of consensus and controversy on the topic of sodium in the heart. The present review focuses on Na(+) channel function and regulation, Na(+) channel structure and function, and Na(+) channel trafficking, sequestration and complexing.


Subject(s)
Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Action Potentials , Amino Acid Sequence , Animals , Congresses as Topic , Humans , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Protein Transport , Sodium Channel Blockers/pharmacology , Sodium Channels/chemistry
19.
Biochim Biophys Acta ; 1833(4): 876-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23103513

ABSTRACT

Essential to beat-to-beat heart function is the ability for cardiomyocytes to propagate electrical excitation and generate contractile force. Both excitation and contractility depend on specific ventricular ion channels, which include the L-type calcium channel (LTCC) and the connexin 43 (Cx43) gap junction. Each of these two channels is localized to a distinct subdomain of the cardiomyocyte plasma membrane. In this review, we focus on regulatory mechanisms that govern the lifecycles of LTCC and Cx43, from their biogenesis in the nucleus to directed delivery to T-tubules and intercalated discs, respectively. We discuss recent findings on how alternative promoter usage, tissue-specific transcription, and alternative splicing determine precise ion channel expression levels within a cardiomyocyte. Moreover, recent work on microtubule and actin-dependent trafficking for Cx43 and LTCC are introduced. Lastly, we discuss how human cardiac disease phenotypes can be attributed to defects in distinct mechanisms of channel regulation at the level of gene expression and channel trafficking. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Subject(s)
Calcium Channels, L-Type/genetics , Connexin 43/genetics , Gene Expression Regulation , Heart Failure/genetics , Myocardium/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing , Calcium Channels, L-Type/metabolism , Cell Nucleus/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Gap Junctions/pathology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Mutation , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
SELECTION OF CITATIONS
SEARCH DETAIL