Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article En | MEDLINE | ID: mdl-33785600

The contraction of heart cells is controlled by the intermolecular signaling between L-type Ca2+ channels (LCCs) and ryanodine receptors (RyRs), and the nanodistance between them depends on the interaction between junctophilin-2 (JPH2) in the sarcoplasmic reticulum (SR) and caveolin-3 (CAV3) in the transversal tubule (TT). In heart failure, decreased expression of JPH2 compromises LCC-RyR communication leading to deficient blood-pumping power. In the present study, we found that JPH2 and CAV3 transcription was concurrently regulated by serum response factor (SRF) and myocardin. In cardiomyocytes from torpid ground squirrels, compared with those from euthermic counterparts, myocardin expression was up-regulated, which boosted both JPH2 and CAV3 expression. Transmission electron microscopic imaging showed that the physical coupling between TTs and SRs was tightened during hibernation and after myocardin overexpression. Confocal Ca2+ imaging under the whole-cell patch clamp condition revealed that these changes enhanced the efficiency of LCC-RyR intermolecular signaling and fully compensated the adaptive down-regulation of LCCs, maintaining the power of heart contraction while avoiding the risk of calcium overload during hibernation. Our finding not only revealed an essential molecular mechanism underlying the survival of hibernating mammals, but also demonstrated a "reverse model of heart failure" at the molecular level, suggesting a strategy for treating heart diseases.


Calcium Signaling , Hibernation , Myocytes, Cardiac/metabolism , Animals , Caveolins/genetics , Caveolins/metabolism , Cells, Cultured , Excitation Contraction Coupling , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/blood , Nuclear Proteins/metabolism , Sciuridae , Trans-Activators/blood , Trans-Activators/metabolism
2.
Med Image Anal ; 70: 102002, 2021 05.
Article En | MEDLINE | ID: mdl-33657508

The Endoscopy Computer Vision Challenge (EndoCV) is a crowd-sourcing initiative to address eminent problems in developing reliable computer aided detection and diagnosis endoscopy systems and suggest a pathway for clinical translation of technologies. Whilst endoscopy is a widely used diagnostic and treatment tool for hollow-organs, there are several core challenges often faced by endoscopists, mainly: 1) presence of multi-class artefacts that hinder their visual interpretation, and 2) difficulty in identifying subtle precancerous precursors and cancer abnormalities. Artefacts often affect the robustness of deep learning methods applied to the gastrointestinal tract organs as they can be confused with tissue of interest. EndoCV2020 challenges are designed to address research questions in these remits. In this paper, we present a summary of methods developed by the top 17 teams and provide an objective comparison of state-of-the-art methods and methods designed by the participants for two sub-challenges: i) artefact detection and segmentation (EAD2020), and ii) disease detection and segmentation (EDD2020). Multi-center, multi-organ, multi-class, and multi-modal clinical endoscopy datasets were compiled for both EAD2020 and EDD2020 sub-challenges. The out-of-sample generalization ability of detection algorithms was also evaluated. Whilst most teams focused on accuracy improvements, only a few methods hold credibility for clinical usability. The best performing teams provided solutions to tackle class imbalance, and variabilities in size, origin, modality and occurrences by exploring data augmentation, data fusion, and optimal class thresholding techniques.


Artifacts , Deep Learning , Algorithms , Endoscopy, Gastrointestinal , Humans
3.
Cardiovasc Res ; 116(13): 2069-2080, 2020 11 01.
Article En | MEDLINE | ID: mdl-32031586

AIMS: ß-adrenergic receptors (ßARs) play pivotal roles in regulating cardiac excitation-contraction (E-C) coupling. Global signalling of ß1ARs up-regulates both the influx of Ca2+ through sarcolemmal L-type Ca2+ channels (LCCs) and the release of Ca2+ from the sarcoplasmic reticulum (SR) through the ryanodine receptors (RyRs). However, we recently found that ß2AR stimulation meditates 'offside compartmentalization', confining ß1AR signalling into subsarcolemmal nanodomains without reaching SR proteins. In the present study, we aim to investigate the new question, whether and how compartmentalized ß1AR signalling regulates cardiac E-C coupling. METHODS AND RESULTS: By combining confocal Ca2+ imaging and patch-clamp techniques, we investigated the effects of compartmentalized ßAR signalling on E-C coupling at both cellular and molecular levels. We found that simultaneous activation of ß2 and ß1ARs, in contrast to global signalling of ß1ARs, modulated neither the amplitude and spatiotemporal properties of Ca2+ sparks nor the kinetics of the RyR response to LCC Ca2+ sparklets. Nevertheless, by up-regulating LCC current, compartmentalized ß1AR signalling synchronized RyR Ca2+ release and increased the functional reserve (stability margin) of E-C coupling. In circumstances of briefer excitation durations or lower RyR responsivity, compartmentalized ßAR signalling, by increasing the intensity of Ca2+ triggers, helped stabilize the performance of E-C coupling and enhanced the Ca2+ transient amplitude in failing heart cells. CONCLUSION: Given that compartmentalized ßAR signalling can be induced by stress-associated levels of catecholamines, our results revealed an important, yet unappreciated, heart regulation mechanism that is autoadaptive to varied stress conditions.


Calcium Channels, L-Type/metabolism , Calcium Signaling , Cardiomegaly/metabolism , Excitation Contraction Coupling , Myocardial Contraction , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-1/metabolism , Action Potentials , Adrenergic Agonists/pharmacology , Animals , Calcium Signaling/drug effects , Cardiomegaly/physiopathology , Computer Simulation , Disease Models, Animal , Excitation Contraction Coupling/drug effects , Kinetics , Male , Microscopy, Confocal , Models, Cardiovascular , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
4.
Cardiovasc Res ; 113(3): 332-342, 2017 03 01.
Article En | MEDLINE | ID: mdl-28077437

Aims: The heart contraction is controlled by the Ca2+-induced Ca2+ release (CICR) between L-type Ca2+ channels and ryanodine receptors (RyRs). The FK506-binding protein FKBP12.6 binds to RyR subunits, but its role in stabilizing RyR function has been debated for long. Recent reports of high-resolution RyR structure show that the HD2 domain that binds to the SPRY2 domain of neighbouring subunit in FKBP-bound RyR1 is detached and invisible in FKBP-null RyR2. The present study was to test the consequence of FKBP12.6 absence on the in situ activation of RyR2. Methods and results: Using whole-cell patch-clamp combined with confocal imaging, we applied a near threshold depolarization to activate a very small fraction of LCCs, which in turn activated RyR Ca2+ sparks stochastically. FKBP12.6-knockout and FK506/rapamycin treatments increased spark frequency and LCC-RyR coupling fidelity without altering LCC open probability. Neither FK506 nor rapamycin further altered LCC-RyR coupling fidelity in FKBP12.6-knockout cells. In loose-seal patch-clamp experiments, the LCC-RyR signalling kinetics, indexed by the delay for a LCC sparklet to trigger a RyR spark, was accelerated after FKBP12.6 knockout and FK506/rapamycin treatments. These results demonstrated that RyRs became more sensitive to Ca2+ triggers without FKBP12.6. Isoproterenol (1 µM) further accelerated the LCC-RyR signalling in FKBP12.6-knockout cells. The synergistic sensitization of RyRs by catecholaminergic signalling and FKBP12.6 dysfunction destabilized the CICR system, leading to chaotic Ca2+ waves and ventricular arrhythmias. Conclusion: FKBP12.6 keeps the RyRs from over-sensitization, stabilizes the potentially regenerative CICR system, and thus may suppress the life-threatening arrhythmogenesis.


Arrhythmias, Cardiac/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling , Myocytes, Cardiac/metabolism , Receptor Cross-Talk , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Proteins/deficiency , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Genotype , Isoproterenol/pharmacology , Kinetics , Male , Membrane Potentials , Mice, Knockout , Microscopy, Confocal , Models, Molecular , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Receptor Cross-Talk/drug effects , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/drug effects , Sirolimus/pharmacology , Stochastic Processes , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/antagonists & inhibitors , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/genetics
5.
Sci Bull (Beijing) ; 62(19): 1295-1303, 2017 Oct 15.
Article En | MEDLINE | ID: mdl-36659291

The elementary Ca2+ release events, Ca2+ sparks, has been found for a quarter of century. However, the molecular regulation of the spark generator, the ryanodine receptor (RyR) on the sarcoplasmic reticulum, remains obscure. Although each subunit of the RyR homotetramer has a site for FK506-binding protein (FKBP), the role of FKBPs in modifying RyR Ca2+ sparks has been debated for long. One of the reasons behind the controversy is that most previous studies detect spontaneous sparks, where the mixture with out-of-focus events and local wavelets prevents an accurate characterization of Ca2+ sparks. In the present study, we detected Ca2+ sparks triggered by single L-type Ca2+ channels (LCCs) under loose-seal patch clamp conditions in FK506-treated or FKBP12.6 knockout cardiomyocytes. We found that FKBP dissociation both by FK506 and by rapamycin decreased the Ca2+ spark amplitude in ventricular cardiomyocytes. This change was neither due to decreased releasable Ca2+ in the sarcoplasmic reticulum, nor explained by changed RyR sensitivity. Actually FK506 increased the LCC-RyR coupling probability and curtailed the latency for an LCC to trigger a RyR Ca2+ spark. FKBP12.6 knockout had similar effects as FK506/rapamycin treatment, indicating that the decreased spark amplitude was attributable to the dissociation of FKBP12.6 rather than FKBP12. We also explained how decreased amplitude of spontaneous sparks after FKBP dissociation sometimes appears to be increased or unchanged due to inappropriate data processing. Our results provided firm evidence that without the inter-RyR coordination by functional FKBP12.6, the RyR recruitment during a Ca2+ spark would be compromised despite the sensitization of individual RyRs.

6.
Med Image Anal ; 35: 489-502, 2017 01.
Article En | MEDLINE | ID: mdl-27614792

Colorectal adenocarcinoma originating in intestinal glandular structures is the most common form of colon cancer. In clinical practice, the morphology of intestinal glands, including architectural appearance and glandular formation, is used by pathologists to inform prognosis and plan the treatment of individual patients. However, achieving good inter-observer as well as intra-observer reproducibility of cancer grading is still a major challenge in modern pathology. An automated approach which quantifies the morphology of glands is a solution to the problem. This paper provides an overview to the Gland Segmentation in Colon Histology Images Challenge Contest (GlaS) held at MICCAI'2015. Details of the challenge, including organization, dataset and evaluation criteria, are presented, along with the method descriptions and evaluation results from the top performing methods.


Algorithms , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/pathology , Diagnostic Imaging/methods , Histological Techniques , Automation , Datasets as Topic , Humans , Reproducibility of Results
7.
Circ Res ; 112(4): 601-5, 2013 Feb 15.
Article En | MEDLINE | ID: mdl-23307820

RATIONALE: During the transition from compensated hypertrophy to heart failure, the signaling between L-type Ca(2+) channels in the cell membrane/T-tubules and ryanodine receptors in the sarcoplasmic reticulum becomes defective, partially because of the decreased expression of a T-tubule-sarcoplasmic reticulum anchoring protein, junctophilin-2. MicroRNA (miR)-24, a junctophilin-2 suppressing miR, is upregulated in hypertrophied and failing cardiomyocytes. OBJECTIVE: To test whether miR-24 suppression can protect the structural and functional integrity of L-type Ca(2+) channel-ryanodine receptor signaling in hypertrophied cardiomyocytes. METHODS AND RESULTS: In vivo silencing of miR-24 by a specific antagomir in an aorta-constricted mouse model effectively prevented the degradation of heart contraction, but not ventricular hypertrophy. Electrophysiology and confocal imaging studies showed that antagomir treatment prevented the decreases in L-type Ca(2+) channel-ryanodine receptor signaling fidelity/efficiency and whole-cell Ca(2+) transients. Further studies showed that antagomir treatment stabilized junctophilin-2 expression and protected the ultrastructure of T-tubule-sarcoplasmic reticulum junctions from disruption. CONCLUSIONS: MiR-24 suppression prevented the transition from compensated hypertrophy to decompensated hypertrophy, providing a potential strategy for early treatment against heart failure.


Calcium Signaling/drug effects , Excitation Contraction Coupling/drug effects , Heart Failure/prevention & control , Hypertrophy, Left Ventricular/drug therapy , MicroRNAs/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Oligonucleotides, Antisense/therapeutic use , Animals , Aortic Stenosis, Subvalvular/complications , Calcium Channels, L-Type/physiology , Calcium Signaling/physiology , Disease Progression , Drug Evaluation, Preclinical , Gene Expression Regulation , Heart Failure/etiology , Heart Failure/metabolism , Hypertrophy, Left Ventricular/complications , Hypertrophy, Left Ventricular/physiopathology , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/physiology , Models, Cardiovascular , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Oligonucleotides, Antisense/pharmacology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/physiology , Sarcoplasmic Reticulum/ultrastructure
8.
Circ Res ; 111(7): 837-41, 2012 Sep 14.
Article En | MEDLINE | ID: mdl-22891046

RATIONALE: Failing cardiomyocytes exhibit decreased efficiency of excitation-contraction (E-C) coupling. The downregulation of junctophilin-2 (JP2), a protein anchoring the sarcoplasmic reticulum to T-tubules, has been identified as a major mechanism underlying the defective E-C coupling. However, the regulatory mechanism of JP2 remains unknown. OBJECTIVE: To determine whether microRNAs regulate JP2 expression. METHODS AND RESULTS: Bioinformatic analysis predicted 2 potential binding sites of miR-24 in the 3'-untranslated regions of JP2 mRNA. Luciferase assays confirmed that miR-24 suppressed JP2 expression by binding to either of these sites. In the aortic stenosis model, miR-24 was upregulated in failing cardiomyocytes. Adenovirus-directed overexpression of miR-24 in cardiomyocytes decreased JP2 expression and reduced Ca(2+) transient amplitude and E-C coupling gain. CONCLUSIONS: MiR-24-mediated suppression of JP2 expression provides a novel molecular mechanism for E-C coupling regulation in heart cells and suggests a new target against heart failure.


Aortic Valve Stenosis/metabolism , Heart Failure/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Up-Regulation , Animals , Aortic Valve Stenosis/pathology , Calcium/metabolism , Cells, Cultured , Computational Biology , Excitation Contraction Coupling/physiology , Heart Failure/pathology , Membrane Proteins/genetics , MicroRNAs/genetics , Models, Animal , Myocytes, Cardiac/pathology , RNA, Messenger/metabolism , Rats , Sarcoplasmic Reticulum/physiology
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 31(4): 610-4, 2011 Apr.
Article Zh | MEDLINE | ID: mdl-21515453

OBJECTIVE: To design a platform for microarray data analysis and processing in the browser/server mode running in Linux operating system. METHODS: Based on the Apache HTTP server, the platform, programmed with Perl language, integrated R language and Bioconductor packages for processing and analysis of the input data of oligonucleotide arrays and two-color spotted arrays. Users were allowed to submit data and parameter configurations to the platform via the web page, and the results of analysis were also returned via the web page. RESULTS: With an easy operation and high performance, the platform fulfilled the functions of processing, quality assessment, biological annotation and statistical analysis of the data from oligonucleotide arrays and two-color spotted arrays. Using the platform, we analyzed the gene expression profiles in Mtb-stimulated macrophages of three clinical phenotypes, namely latent TB (LTB), pulmonary (PTB) and meningeal (TBM), and obtained valuable clues for identifying tuberculosis susceptibility genes. We also analyzed the effect of INH treatment on Mycobacterium tuberculosis gene expression in various dormancy models, such as hypoxia and KatG mutant, and found that a set of genes responded to INH treatment during exponential growth but not in dormancy, and that the overall number of differentially regulated genes was reduced in the cells in low metabolic state. CONCLUSION: The platform we have constructed integrates comprehensive resources, and with a user-friendly interface, allows direct result visualization to facilitate microarray data analysis.


Computational Biology/methods , Oligonucleotide Array Sequence Analysis , Software , Databases, Genetic , Gene Expression Profiling/methods , Genetic Predisposition to Disease , Humans , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/genetics , Tuberculosis/genetics , Tuberculosis/immunology , User-Computer Interface
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(2): 219-23, 2010 Feb.
Article Zh | MEDLINE | ID: mdl-20159684

OBJECTIVE: To design a versatile genome comparison and visualization platform based on browser/server mode supported by a local server. METHODS: The server of the platform was Apache HTTP server. Perl was used to integrate such genome alignment package and algorithms as MUMmer, LAGAN, and Mauve for different comparison purposes, and the users could submit data and parameters to the platform via the web page. The results of analysis were also returned via the web page. RESULTS: The platform could handle multiple data input formats, compare complete and draft genome sequence, alignment pair-wise or multi genome of more divergent species, identify regions of high similarity, locate local nucleotide mutations and large-scale recombination, and display the results by visualization interface. Analysis of the homology of 10 new strains of influenza A virus indicated that PB1 gene might evolve from human H3N2 viruses, PB2 and PA genes from avian H3N2 viruses, and HA and NS genes from swine H1N1 viruses. Alignment of Mycobacterium tuberculosis (H37Rv, CDC1551) and Mycobacterium bovis (AF2122/97) genomes revealed that sequence insertion/deletion and duplication were the major source of genomic differences. CONCLUSION: The platform integrate comprehensive resources with a user-friendly interface and intuitive result visualization to facilitate conventional study of comparative genomics.


Genome , Genomics/methods , Software , Base Sequence , Comparative Genomic Hybridization/methods , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Molecular Sequence Data , Mycobacterium tuberculosis/genetics , User-Computer Interface
11.
Proc Natl Acad Sci U S A ; 106(42): 18028-33, 2009 Oct 20.
Article En | MEDLINE | ID: mdl-19815510

As the most prototypical G protein-coupled receptor, beta-adrenergic receptor (betaAR) regulates the pace and strength of heart beating by enhancing and synchronizing L-type channel (LCC) Ca(2+) influx, which in turn elicits greater sarcoplasmic reticulum (SR) Ca(2+) release flux via ryanodine receptors (RyRs). However, whether and how betaAR-protein kinase A (PKA) signaling directly modulates RyR function remains elusive and highly controversial. By using unique single-channel Ca(2+) imaging technology, we measured the response of a single RyR Ca(2+) release unit, in the form of a Ca(2+) spark, to its native trigger, the Ca(2+) sparklet from a single LCC. We found that acute application of the selective betaAR agonist isoproterenol (1 microM, < or = 20 min) increased triggered spark amplitude in an LCC unitary current-independent manner. The increased ratio of Ca(2+) release flux underlying a Ca(2+) spark to SR Ca(2+) content indicated that betaAR stimulation helps to recruit additional RyRs in synchrony. Quantification of sparklet-spark kinetics showed that betaAR stimulation synchronized the stochastic latency and increased the fidelity (i.e., chance of hit) of LCC-RyR intermolecular signaling. The RyR modulation was independent of the increased SR Ca(2+) content. The PKA antagonists Rp-8-CPT-cAMP (100 microM) and H89 (10 microM) both eliminated these effects, indicating that betaAR acutely modulates RyR activation via the PKA pathway. These results demonstrate unequivocally that RyR activation by a single LCC is accelerated and synchronized during betaAR stimulation. This molecular mechanism of sympathetic regulation will permit more fundamental studies of altered betaAR effects in cardiovascular diseases.


Calcium Channels, L-Type/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , In Vitro Techniques , Isoproterenol/pharmacology , Microscopy, Confocal , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/metabolism , Signal Transduction/physiology
12.
Di Yi Jun Yi Da Xue Xue Bao ; 25(4): 413-5, 2005 Apr.
Article Zh | MEDLINE | ID: mdl-15837641

OBJECTIVE: To establish a method by high-performance liquid chromatography (HPLC) for determining the concentration of magnetic mitomycin C-polybutylcyanoacrylate nanoparticles in mouse plasma. METHODS: Chromatography was performed on a LiChroCART C18 (250 mm x 4 mm, 5 microm) column with the mobile phase consisting of acetonitrile-NaAC (15:85), the flow rate of 1.0 ml/min, and the detection wavelength of 365 nm. Sample extraction was carried out with ethylacetate. RESULTS: The linear range of mouse plasma mitomycin C concentration was 0.04-1.00 microg/ml, and the linear equation of Y=16 388X-17.17 (r=0.999 8) was derived. CONCLUSION: This method is very easy to operate and suits the need of perclinical pharmacokinetic studies of mitomycin-magnetic nanoparticles and yields accurate and precise results.


Drug Delivery Systems , Enbucrilate , Mitomycin/blood , Nanoparticles , Animals , Chromatography, High Pressure Liquid , Drug Carriers , Magnetics , Mice , Mitomycin/administration & dosage
...