Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
bioRxiv ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38659858

ABSTRACT

Natural Killer (NK) cells can recognize and kill Mtb-infected cells in vitro, however their role after natural human exposure has not been well-studied. To identify Mtb-responsive NK cell populations, we analyzed the peripheral blood of healthy household contacts of active Tuberculosis (TB) cases and source community donors in an endemic region of Port-au-Prince, Haiti by flow cytometry. We observed higher CD8α expression on NK cells in putative resistors (IGRA- contacts) with a progressive loss of these circulating cells during household-associated latent infection and disease. In vitro assays and CITE-seq analysis of CD8α+ NK cells demonstrated enhanced maturity, cytotoxic gene expression, and response to cytokine stimulation relative to CD8α- NK cells. CD8α+ NK cells also displayed dynamic surface expression dependent on MHC I in contrast to conventional CD8+ T cells. Together, these results support a specialized role for CD8α+ NK cell populations during Mtb infection correlating with disease resistance.

2.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-37546948

ABSTRACT

Most human pancreatic ductal adenocarcinoma (PDAC) are not infiltrated with cytotoxic T cells and are highly resistant to immunotherapy. Over 90% of PDAC have oncogenic KRAS mutations, and phosphoinositide 3-kinases (PI3Ks) are direct effectors of KRAS. Our previous study demonstrated that ablation of Pik3ca in KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cells induced host T cells to infiltrate and completely eliminate the tumors in a syngeneic orthotopic implantation mouse model. Now, we show that implantation of Pik3ca-/- KPC (named αKO) cancer cells induces clonal expansion of cytotoxic T cells infiltrating the pancreatic tumors. To identify potential molecules that can regulate the activity of these anti-tumor T cells, we conducted an in vivo genome-wide gene-deletion screen using αKO cells implanted in the mouse pancreas. The result shows that deletion of propionyl-CoA carboxylase subunit B gene (Pccb) in αKO cells (named p-αKO) leads to immune evasion, tumor progression and death of host mice. Surprisingly, p-αKO tumors are still infiltrated with clonally expanded CD8+ T cells but they are inactive against tumor cells. However, blockade of PD-L1/PD1 interaction reactivated these clonally expanded T cells infiltrating p-αKO tumors, leading to slower tumor progression and improve survival of host mice. These results indicate that Pccb can modulate the activity of cytotoxic T cells infiltrating some pancreatic cancers and this understanding may lead to improvement in immunotherapy for this difficult-to-treat cancer.

3.
Invest Ophthalmol Vis Sci ; 64(13): 37, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37870847

ABSTRACT

Purpose: The lens epithelium maintains the overall health of the organ. We used single-cell RNA sequencing (scRNA-seq) technology to assess transcriptional heterogeneity between cells in the postnatal day 2 (P2) epithelium and identify distinct epithelial cell subtypes. Analysis of these data was used to better understand lens growth, differentiation, and homeostasis on P2. Methods: scRNA-seq on P2 mouse lenses was performed using the 10x Genomics Chromium Single Cell 3' Kit (v3.1) and short-read Illumina sequencing. Sequence alignment and preprocessing of data were conducted using 10x Genomics Cell Ranger software. Seurat was employed for preprocessing, quality control, dimensionality reduction, and cell clustering, and Monocle was utilized for trajectory analysis to understand the developmental progression of the lens cells. CellChat and GO analyses were used to explore cell-cell communication networks and signaling interactions. Results: Lens epithelial cells (LECs) were divided into seven subclusters, classified by specific gene markers. The expression of crystallin, cell-cycle, and metabolic genes was not uniform, indicating distinct functional roles of LECs. Trajectory analysis predicted a bifurcation of differentiating and cycling cells from an Igfbp5+ progenitor pool. We also identified heterogeneity in signaling molecules and pathways, suggesting that cycling and progenitor subclusters have prominent roles in coordinating crosstalk. Conclusions: scRNA-seq corroborated many known markers of epithelial differentiation and proliferation while providing further insight into the pathways and genes directing these processes. Interestingly, we demonstrated that the developing epithelium can be divided into distinct subpopulations. These clusters reflect the transcriptionally diverse roles of the epithelium in proliferation, signaling, and maintenance.


Subject(s)
Lens, Crystalline , Animals , Mice , Lens, Crystalline/metabolism , Epithelium , Epithelial Cells/metabolism , Cell Differentiation , Sequence Analysis, RNA
4.
Sleep Health ; 7(5): 543-547, 2021 10.
Article in English | MEDLINE | ID: mdl-34417148

ABSTRACT

OBJECTIVES: To investigate adolescent sleep parameters and predictors during COVID-19-related school closures. METHODS: Original data were analyzed from a cross-sectional online survey of 590 teens in grades 6-12 attending school remotely in 35 US states, in May/June 2020. RESULTS: Students reported waking up 2.1-2.9 hours later during school closures and averaged 7.9-8.7 hours of sleep and 8.6-9.5 hours in bed on school nights. Compared to middle schoolers, high school students had later bed and wake times, accompanied by spending less time in bed and less time sleeping. The delay in wake time after school closures was also longer for high school students than for middle schoolers. Students with later class start times went to bed later, but also woke up later, slept longer, and spent more time in bed. When comparing intraindividual sleep before and after school closures, later class start times resulted in greater delays in wake time and greater odds of increased sleep duration. In addition, parent-set bedtimes were associated with earlier bedtimes and longer sleep duration during school closures. CONCLUSIONS: As a result of COVID-19-related school closures and remote instruction, more middle and high school students achieved recommended amounts of sleep, primarily by waking up later in the morning. This study supports previous evidence that morning start schedule affects adolescent sleep behaviors. The implications of this study extend beyond COVID-19 school closures; adolescent sleep health improves with later school start times and fewer scheduled morning activities.


Subject(s)
COVID-19 , Adolescent , Cross-Sectional Studies , Humans , SARS-CoV-2 , Schools , Sleep
5.
FASEB J ; 34(9): 12663-12676, 2020 09.
Article in English | MEDLINE | ID: mdl-32761888

ABSTRACT

Phospholipase Cß1 is activated by Gαq to generate calcium signals in response to hormones and neurotransmitters. Besides carrying out this plasma membrane function, PLCß1 has a cytosolic population that helps to drive the differentiation of PC12 cells by inhibiting a nuclease that promotes RNA-induced silencing (C3PO). Here, we show that down-regulating PLCß1 or reducing its cytosolic population by activating Gαq to localize it to the plasma membrane returns differentiated PC12 and SK-N-SH cells to an undifferentiated state. In this state, PC12 cells have a spherical morphology, resume proliferation, and express the stem cell transcription factors nanog and Oct4. Similar changes are seen when C3PO is down-regulated. This return to a stem-like state is accompanied by shifts in multiple miR populations. Surprisingly, de-differentiation can be induced by extended stimulation of Gαq where cells return to a spherical morphology and levels of specific miRs return to their undifferentiated values. In complementary studies, we followed the real-time hydrolysis of a fluorescent-tagged miR in cells where PLCß1 or C3PO were down-regulated in PC12 cells and find substantial differences in miR processing in the undifferentiated and differentiated states. Taken together, our studies suggest that PLCß1, through its ability to regulate C3PO and endogenous miR populations, mediates the differentiation of two types of cultured neuronal cells.


Subject(s)
Cell Dedifferentiation , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phospholipase C beta/metabolism , RNA-Induced Silencing Complex/metabolism , Animals , Cell Line, Tumor , Humans , MicroRNAs/metabolism , PC12 Cells , RNA Interference , Rats , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 117(7): 3858-3866, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32015129

ABSTRACT

The accepted role of the protein Kv2.1 in arterial smooth muscle cells is to form K+ channels in the sarcolemma. Opening of Kv2.1 channels causes membrane hyperpolarization, which decreases the activity of L-type CaV1.2 channels, lowering intracellular Ca2+ ([Ca2+]i) and causing smooth muscle relaxation. A limitation of this model is that it is based exclusively on data from male arterial myocytes. Here, we used a combination of electrophysiology as well as imaging approaches to investigate the role of Kv2.1 channels in male and female arterial myocytes. We confirmed that Kv2.1 plays a canonical conductive role but found it also has a structural role in arterial myocytes to enhance clustering of CaV1.2 channels. Less than 1% of Kv2.1 channels are conductive and induce membrane hyperpolarization. Paradoxically, by enhancing the structural clustering and probability of CaV1.2-CaV1.2 interactions within these clusters, Kv2.1 increases Ca2+ influx. These functional impacts of Kv2.1 depend on its level of expression, which varies with sex. In female myocytes, where expression of Kv2.1 protein is higher than in male myocytes, Kv2.1 has conductive and structural roles. Female myocytes have larger CaV1.2 clusters, larger [Ca2+]i, and larger myogenic tone than male myocytes. In contrast, in male myocytes, Kv2.1 channels regulate membrane potential but not CaV1.2 channel clustering. We propose a model in which Kv2.1 function varies with sex: in males, Kv2.1 channels control membrane potential but, in female myocytes, Kv2.1 plays dual electrical and CaV1.2 clustering roles. This contributes to sex-specific regulation of excitability, [Ca2+]i, and myogenic tone in arterial myocytes.


Subject(s)
Arteries/metabolism , Calcium Channels, L-Type/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Shab Potassium Channels/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Cells, Cultured , Female , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Shab Potassium Channels/genetics
7.
Transl Psychiatry ; 8(1): 165, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30139969

ABSTRACT

The effect of six different traumatic stress protocols on the transcriptome of the rat adrenal gland was examined using RNA sequencing. These protocols included chronic variable stress, chronic shock, social defeat and social isolation. The response of the transcriptome to stress suggested that there are genes that respond in a universal or stress modality-independent manner, as well as genes that respond in a stress modality-specific manner. Using a small number of the genes selected from the modality-independent set of stress-sensitive genes, a sensitive and robust measure of chronic stress exposure was developed. This stress-sensitive gene expression (SSGE) index could detect chronic traumatic stress exposure in a wide range of different stress models in a manner that was relatively independent of the modality of stress exposure and that paralleled the intensity of stress exposure in a dose-dependent manner. This measure could reliably distinguish control and stressed individuals in the case of animals exposed to the most intense stress protocols. The response of a subset of the modality-specific genes could also distinguish some types of stress exposure, based solely on changes in the pattern of gene expression. The results suggest that it is possible to develop diagnostic measures of traumatic stress exposure based solely on changes in the level of expression of a relatively small number of genes.


Subject(s)
Stress, Psychological/genetics , Stress, Psychological/psychology , Transcriptome , Animals , Behavior Rating Scale , Behavior, Animal , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA
8.
Prog Biophys Mol Biol ; 122(3): 165-186, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27702655

ABSTRACT

Evolution has acted to shape the action potential in different regions of the heart in order to produce a maximally stable and efficient pump. This has been achieved by creating regional differences in ion channel expression levels within the heart as well as differences between equivalent cardiac tissues in different species. These region- and species-dependent differences in channel expression are established by regulatory evolution, evolution of the regulatory mechanisms that control channel expression levels. Ion channel auxiliary subunits are obvious targets for regulatory evolution, in order to change channel expression levels and/or modify channel function. This review focuses on the transmural gradients of ion channel expression in the heart and the role that regulation of auxiliary subunit expression plays in generating and shaping these gradients.


Subject(s)
Gene Expression Regulation , Ion Channels/genetics , Ion Channels/metabolism , Protein Subunits/genetics , Animals , Electrophysiological Phenomena , Humans , Potassium/metabolism
9.
Adv Biol Regul ; 61: 51-7, 2016 05.
Article in English | MEDLINE | ID: mdl-26746047

ABSTRACT

Phosphoinositide-specific-phospholipase Cß (PLCß) is the main effector of Gαq stimulation which is coupled to receptors that bind acetylcholine, bradykinin, dopamine, angiotensin II as well as other hormones and neurotransmitters. Using a yeast two-hybrid and other approaches, we have recently found that the same region of PLCß that binds Gαq also interacts with Component 3 Promoter of RNA induced silencing complex (C3PO), which is required for efficient activity of the RNA-induced silencing complex. In purified form, C3PO competes with Gαq for PLCß binding and at high concentrations can quench PLCß activation. Additionally, we have found that the binding of PLCß to C3PO inhibits its nuclease activity leading to reversal of RNA-induced silencing of specific genes. In cells, we found that PLCß distributes between the plasma membrane where it localizes with Gαq, and in the cytosol where it localizes with C3PO. When cells are actively processing small interfering RNAs the interaction between PLCß and C3PO gets stronger and leads to changes in the cellular distribution of PLCß. The magnitude of attenuation is specific for different silencing RNAs. Our studies imply a direct link between calcium responses mediated through Gαq and post-transcriptional gene regulation through PLCß.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Phospholipase C beta/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA-Induced Silencing Complex/genetics , Signal Transduction/genetics , Animals , Binding Sites , Binding, Competitive , Calcium/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Phospholipase C beta/metabolism , Protein Binding , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/metabolism
10.
PLoS One ; 10(4): e0120785, 2015.
Article in English | MEDLINE | ID: mdl-25875483

ABSTRACT

It has been suggested that optimization of either axonal conduction velocity or the energy efficiency of action potential conduction predominates in the selection of voltage-gated sodium conductance levels in the squid axon. A population genetics model of channel gene regulatory function was used to examine the role of these and other evolutionary forces on the selection of both sodium and potassium channel expression levels. In this model, the accumulating effects of mutations result in degradation of gene regulatory function, causing channel gene expression to fall to near-zero in the absence of positive selection. In the presence of positive selection, channel expression levels fall to the lowest values consistent with the selection criteria, thereby establishing a selection-mutation balance. Within the parameter space of sodium and potassium conductance values, the physiological performance of the squid axon model showed marked discontinuities associated with conduction failure and excitability. These discontinuities in physiological function may produce fitness cliffs. A fitness cliff associated with conduction failure, combined with the effects of phenotypic noise, can account for the selection of sodium conductance levels, without considering either conduction velocity or metabolic cost. A fitness cliff associated with a transition in axonal excitability, combined with phenotypic noise, can explain the selection of potassium channel expression levels. The results suggest that voltage-gated ion channel expression will fall to low levels, consistent with key functional constraints, even in the absence of positive selection for energy efficiency. Channel expression levels and individual variation in channel expression within the population can be explained by regulatory evolution in combination with genetic variation in regulatory function and phenotypic noise, without resorting to more complex mechanisms, such as activity-dependent homeostasis. Only a relatively small region of the large, nominally isofunctional parameter space for channel expression will normally be occupied, because of the effects of mutation.


Subject(s)
Axons/physiology , Biological Evolution , Decapodiformes/physiology , Potassium Channels/genetics , Sodium Channels/genetics , Action Potentials , Animals , Decapodiformes/genetics , Evolution, Molecular , Gene Regulatory Networks , Models, Genetic , Mutation , Neural Conduction , Potassium Channels/metabolism , Sodium Channels/metabolism
11.
Front Pharmacol ; 6: 9, 2015.
Article in English | MEDLINE | ID: mdl-25691868

ABSTRACT

4-phenylbutyrate (4-PB) has been shown to increase the protein content in a number of cells types. One such protein is Connexin43 (Cx43). We show here that 4-phenylbutyrate exposure results in significantly elevated cell to cell coupling, as determined by dual whole cell patch clamp. Incubation with 5 mM 4PB for 24 h or more nearly doubles junctional conductance. Interestingly, mRNA levels for Cx43 declined with exposure to 4-PB while western blot analysis revealed not significant change in protein levels. These data are most consistent with stabilization of the existing Cx43 pool or alterations in the number of functional channels within an existing pool of active and silent channels. These data represent a baseline for testing the efficacy of increased connexin mediated coupling in a variety of multicellular functions including erectile function.

12.
Proc Natl Acad Sci U S A ; 109(5): 1601-6, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307618

ABSTRACT

Scaling of cardiac electrophysiology with body mass requires large changes in the ventricular action potential duration and heart rate in mammals. These changes in cellular electrophysiological function are produced by systematic and coordinated changes in the expression of multiple ion channel and transporter genes. Expression of one important potassium current, the transient outward current (I(to)), changes significantly during mammalian evolution. Changes in I(to) expression are determined, in part, by variation in the expression of an obligatory auxiliary subunit encoded by the KChIP2 gene. The KChIP2 gene is expressed in both cardiac myocytes and neurons and transcription in both cell types is initiated from the same CpG island promoter. Species-dependent variation of KChIP2 expression in heart is mediated by the evolution of the cis-regulatory function of this gene. Surprisingly, the major locus of evolutionary change for KChIP2 gene expression in heart lies within the CpG island core promoter. The results demonstrate that CpG island promoters are not simply permissive for gene expression but can also contribute to tissue-selective expression and, as such, can function as an important locus for the evolution of cis-regulatory function. More generally, evolution of the cis-regulatory function of voltage-gated ion channel genes appears to be an effective and efficient way to modify channel expression levels to optimize electrophysiological function.


Subject(s)
CpG Islands , Kv Channel-Interacting Proteins/genetics , Myocardium/metabolism , Promoter Regions, Genetic , Amino Acid Sequence , Animals , Base Sequence , DNA Probes , Gene Expression Regulation , Guinea Pigs , Kv Channel-Interacting Proteins/chemistry , Mice , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Sequence Homology, Amino Acid , Transcription, Genetic
13.
Cell Signal ; 24(5): 1109-14, 2012 May.
Article in English | MEDLINE | ID: mdl-22286107

ABSTRACT

α-Synuclein is a conserved protein that is a key component in neurodegenerative plaques [1,2]. α-Synuclein binds strongly to phospholipase Cß (PLCß) and promotes Ca2+ release in cells. Here, we show that expression of α-synuclein increases the cellular level of PLCß1 in two neuronal cell lines: PC12 and SK-N-S-SH. The increase in PLCß1 is not accompanied by changes in the level of RNA or in ubiquitination. Instead, we find that α-synuclein protects PLCß1 from trypsin digestion and from degradation by the Ca(+2) activated protease calpain. Calpain removes the C-terminal region of the enzyme which mediates activation by Gα(q). We find that in SK-N-SH cells, α-synuclein reduced degradation of PLCß1 by calpain during Ca2+ signaling allowing the enzyme to remain sensitive to Gα(q) activation. Taken together, our studies show that α-synuclein protects the integrity of PLCß1 and its ability to be activated by Gα(q), which may in turn impact Ca2+ signaling.


Subject(s)
Phospholipase C beta/metabolism , alpha-Synuclein/physiology , Animals , Calcimycin/pharmacology , Calpain/antagonists & inhibitors , Calpain/chemistry , Calpain/metabolism , Enzyme Activators/pharmacology , HEK293 Cells , Humans , PC12 Cells , Peptide Fragments/chemistry , Phospholipase C beta/chemistry , Phospholipase C beta/genetics , Protein Binding , Proteolysis , Rats , Transcription, Genetic , Trypsin/chemistry , Ubiquitination , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
14.
Circ Arrhythm Electrophysiol ; 4(5): 753-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21828312

ABSTRACT

BACKGROUND: After the recent cloning of light-sensitive ion channels and their expression in mammalian cells, a new field, optogenetics, emerged in neuroscience, allowing for precise perturbations of neural circuits by light. However, functionality of optogenetic tools has not been fully explored outside neuroscience, and a nonviral, nonembryogenesis-based strategy for optogenetics has not been shown before. METHODS AND RESULTS: We demonstrate the utility of optogenetics to cardiac muscle by a tandem cell unit (TCU) strategy, in which nonexcitable cells carry exogenous light-sensitive ion channels, and, when electrically coupled to cardiomyocytes, produce optically excitable heart tissue. A stable channelrhodopsin2 (ChR2)-expressing cell line was developed, characterized, and used as a cell delivery system. The TCU strategy was validated in vitro in cell pairs with adult canine myocytes (for a wide range of coupling strengths) and in cardiac syncytium with neonatal rat cardiomyocytes. For the first time, we combined optical excitation and optical imaging to capture light-triggered muscle contractions and high-resolution propagation maps of light-triggered electric waves, found to be quantitatively indistinguishable from electrically triggered waves. CONCLUSIONS: Our results demonstrate feasibility to control excitation and contraction in cardiac muscle by light, using the TCU approach. Optical pacing in this case uses less energy, offers superior spatiotemporal control and remote access and can serve not only as an elegant tool in arrhythmia research but may form the basis for a new generation of light-driven cardiac pacemakers and muscle actuators. The TCU strategy is extendable to (nonviral) stem cell therapy and is directly relevant to in vivo applications.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Light , Muscle Contraction/physiology , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Communication/physiology , Channelrhodopsins , Coculture Techniques , Dogs , Electric Stimulation , Feasibility Studies , HEK293 Cells , Humans , Kidney/cytology , Kidney/metabolism , Myocytes, Cardiac/cytology , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Transfection
15.
J Physiol ; 589(Pt 13): 3275-88, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21521762

ABSTRACT

Mechanisms that contribute to maintaining expression of functional ion channels at relatively constant levels following perturbations of channel biosynthesis are likely to contribute significantly to the stability of electrophysiological systems in some pathological conditions. In order to examine the robustness of L-type calcium current expression, the response to changes in Ca²âº channel Cav1.2 gene dosage was studied in adult mice. Using a cardiac-specific inducible Cre recombinase system, Cav1.2 mRNA was reduced to 11 ± 1% of control values in homozygous floxed mice and the mice died rapidly (11.9 ± 3 days) after induction of gene deletion. In these homozygous knockout mice, echocardiographic analysis showed that myocardial contractility was reduced to 14 ± 1% of control values shortly before death. For these mice, no effective compensatory changes in ion channel gene expression were triggered following deletion of both Cav1.2 alleles, despite the dramatic decay in cardiac function. In contrast to the homozygote knockout mice, following knockout of only one Cav1.2 allele, cardiac function remained unchanged, as did survival.Cav1.2mRNAexpression in the left ventricle of heterozygous knockout mice was reduced to 58 ± 3% of control values and there was a 21 ± 2% reduction in Cav1.2 protein expression. There was no significant reduction in L-type Ca²âº current density in these mice. The results are consistent with a model of L-type calcium channel biosynthesis in which there are one or more saturated steps, which act to buffer changes in both total Cav1.2 protein and L-type current expression.


Subject(s)
Calcium Channels, L-Type/deficiency , Gene Expression Regulation/genetics , Genetic Carrier Screening , Myocytes, Cardiac/physiology , Age Factors , Alleles , Animals , Calcium Channels, L-Type/biosynthesis , Calcium Channels, L-Type/genetics , Female , Gene Dosage/genetics , Genetic Carrier Screening/methods , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation/genetics
16.
FASEB J ; 25(3): 851-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21084696

ABSTRACT

In vitro models of cardiac hypertrophy focus exclusively on applying "external" dynamic signals (electrical, mechanical, and chemical) to achieve a hypertrophic state. In contrast, here we set out to demonstrate the role of "self-organized" cellular architecture and activity in reprogramming cardiac cell/tissue function toward a hypertrophic phenotype. We report that in neonatal rat cardiomyocyte culture, subtle out-of-plane microtopographic cues alter cell attachment, increase biomechanical stresses, and induce not only structural remodeling, but also yield essential molecular and electrophysiological signatures of hypertrophy. Increased cell size and cell binucleation, molecular up-regulation of released atrial natriuretic peptide, altered expression of classic hypertrophy markers, ion channel remodeling, and corresponding changes in electrophysiological function indicate a state of hypertrophy on par with other in vitro and in vivo models. Clinically used antihypertrophic pharmacological treatments partially reversed hypertrophic behavior in this in vitro model. Partial least-squares regression analysis, combining gene expression and functional data, yielded clear separation of phenotypes (control: cells grown on flat surfaces; hypertrophic: cells grown on quasi-3-dimensional surfaces and treated). In summary, structural surface features can guide cardiac cell attachment, and the subsequent syncytial behavior can facilitate trophic signals, unexpectedly on par with externally applied mechanical, electrical, and chemical stimulation.


Subject(s)
Cardiomegaly , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Adhesion/physiology , Cell Shape/physiology , Cells, Cultured , Electric Stimulation , Genetic Markers , Mechanotransduction, Cellular/physiology , Myocytes, Cardiac/drug effects , Natriuretic Peptide, Brain/pharmacology , Phenotype , Physical Stimulation , Rats , Stimulation, Chemical , Tissue Scaffolds
17.
Evol Dev ; 11(5): 610-8, 2009.
Article in English | MEDLINE | ID: mdl-19754717

ABSTRACT

Cellular electrophysiological systems, like developmental systems, appear to evolve primarily by means of regulatory evolution. It is suggested that electrophysiological systems share two key features with developmental systems that account for this dependence on regulatory evolution. For both systems, structural evolution has the potential to create significant problems of pleiotropy and both systems are predominantly computational in nature. It is concluded that the relative balance of physical and computational tasks that a biological system has to perform, combined with the probability that these tasks may have to change significantly during the course of evolution, will be major factors in determining the relative mix of regulatory and structural evolution that is observed for a given system. Physiological systems that directly interface with the environment will almost always perform some low-level physical task. In the majority of cases this will require evolution of protein function in order for the tasks themselves to evolve. For complex physiological systems a large fraction of their function will be devoted to high-level control functions that are predominantly computational in nature. In most cases regulatory evolution will be sufficient in order for these computational tasks to evolve.


Subject(s)
Cell Physiological Phenomena , Evolution, Molecular , Gene Expression Regulation , Animals , Electrophysiology , Humans
18.
Physiol Genomics ; 35(3): 262-72, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-18765860

ABSTRACT

The relative importance of regulatory versus structural evolution for the evolution of different biological systems is a subject of controversy. The primacy of regulatory evolution in the diversification of morphological traits has been promoted by many evolutionary developmental biologists. For physiological traits, however, the role of regulatory evolution has received less attention or has been considered to be relatively unimportant. To address this issue for electrophysiological systems, we examined the importance of regulatory and structural evolution in the evolution of the electrophysiological function of cardiac myocytes in mammals. In particular, two related phenomena were studied: the change in action potential morphology in small mammals and the scaling of action potential duration across mammalian phylogeny. In general, the functional properties of the ion channels involved in ventricular action potential repolarization were found to be relatively invariant. In contrast, there were large changes in the expression levels of multiple ion channel and transporter genes. For the Kv2.1 and Kv4.2 potassium channel genes, which are primary determinants of the action potential morphology in small mammals, the functional properties of the proximal promoter regions were found to vary in concordance with species-dependent differences in mRNA expression, suggesting that evolution of cis-regulatory elements is the primary determinant of this trait. Scaling of action potential duration was found to be a complex phenomenon, involving changes in the expression of a large number of channels and transporters. In this case, it is concluded that regulatory evolution is the predominant mechanism by which the scaling is achieved.


Subject(s)
Biological Evolution , Electrophysiology/methods , Muscle Cells/physiology , Potassium Channels, Voltage-Gated/physiology , Action Potentials/physiology , Animals , Body Weight , Cattle , Ferrets , Guinea Pigs , Heart Rate , Humans , Mice , Muscle Cells/cytology , Myocardium/cytology , Rabbits , Rats , Species Specificity
19.
J Physiol ; 579(Pt 2): 465-71, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17218348

ABSTRACT

This study examines the molecular basis for the T-type and L-type Ca(2+) currents in canine Purkinje cells. The I(CaT) in Purkinje cells was completely suppressed by 200 nM kurtoxin, a specific blocker of both Ca(v)3.1 and Ca(v)3.2 channels. Since only Ca(v)3.2 mRNA is expressed at high levels in Purkinje fibres, being approximately 100-fold more abundant than either Ca(v)3.1 or Ca(v)3.3 mRNAs, it is concluded that the Ca(v)3.2 gene encodes the bulk of the T-type Ca(2+) channels in canine Purkinje cells. This conclusion is consistent with the sensitivity of the current to blockade by Ni(2+) ions (K(D) = 32 microM). For L-type channels, Ca(v)1.2 mRNA was most abundant in Purkinje fibres but a significant level of Ca(v)1.3 mRNA expression was also found. A comparison of the sensitivity to blockade by isradipine of the L-type currents in Purkinje cells and ventricular epicardial myocytes, which only express Ca(v)1.2, suggests that the Ca(v)1.3 channels make, at most, a minor contribution to the L-type current in canine Purkinje cells.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/physiology , Purkinje Fibers/physiology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/genetics , Dogs , Electrophysiology , Gene Expression Regulation , Isradipine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Scorpion Venoms/pharmacology , Ventricular Function
20.
J Mol Cell Cardiol ; 40(2): 295-302, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16412459

ABSTRACT

Previous studies have shown that regional variation in ion channel gene expression contributes to electrical heterogeneity within the walls of the cardiac ventricles. To map the extent of regional variation in gene expression in the ventricular walls and to begin to understand its genesis we have performed a microarray analysis of gene expression in the epicardial and endocardial tissues of the rat adult left ventricle. While the vast majority of the genes are expressed at uniform levels across the ventricular wall, a total of 36 transcripts (representing less than 0.1% of the genes expressed in the ventricle) are expressed more abundantly in either epicardium or endocardium. One of these differentially expressed genes is the sodium channel gene Scn5a, which is expressed at higher levels in the endocardium than in the epicardium of rat heart. The transcription factor genes Irx3, Irx5 and Etv1 were found to be expressed in transmural gradients across the ventricular wall of rat heart and also of canine heart. The Irx3 and Irx5 genes were expressed in an inverse pattern to that of the Kcnd2 (Kv4.2) gene in rat heart, suggesting that these transcription factors may act as negative regulators of Kcnd2 expression in vivo.


Subject(s)
Myocardium/metabolism , RNA, Messenger/metabolism , Animals , Base Sequence , Endocardium/metabolism , Gene Expression/physiology , Heart Ventricles/metabolism , In Situ Hybridization , Molecular Sequence Data , Pericardium/metabolism , Rats , Sodium Channels/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...