Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27.282
Filter
1.
Europace ; 26(7)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39082711

ABSTRACT

AIMS: Electroanatomical adaptations during the neonatal to adult phase have not been comprehensively studied in preclinical animal models. To explore the impact of age as a biological variable on cardiac electrophysiology, we employed neonatal and adult guinea pigs, which are a recognized animal model for developmental research. METHODS AND RESULTS: Electrocardiogram recordings were collected in vivo from anaesthetized animals. A Langendorff-perfusion system was employed for the optical assessment of action potentials and calcium transients. Optical data sets were analysed using Kairosight 3.0 software. The allometric relationship between heart weight and body weight diminishes with age, it is strongest at the neonatal stage (R2 = 0.84) and abolished in older adults (R2 = 1E-06). Neonatal hearts exhibit circular activation, while adults show prototypical elliptical shapes. Neonatal conduction velocity (40.6 ± 4.0 cm/s) is slower than adults (younger: 61.6 ± 9.3 cm/s; older: 53.6 ± 9.2 cm/s). Neonatal hearts have a longer action potential duration (APD) and exhibit regional heterogeneity (left apex; APD30: 68.6 ± 5.6 ms, left basal; APD30: 62.8 ± 3.6), which was absent in adults. With dynamic pacing, neonatal hearts exhibit a flatter APD restitution slope (APD70: 0.29 ± 0.04) compared with older adults (0.49 ± 0.04). Similar restitution characteristics are observed with extrasystolic pacing, with a flatter slope in neonates (APD70: 0.54 ± 0.1) compared with adults (younger: 0.85 ± 0.4; older: 0.95 ± 0.7). Neonatal hearts display unidirectional excitation-contraction coupling, while adults exhibit bidirectionality. CONCLUSION: Postnatal development is characterized by transient changes in electroanatomical properties. Age-specific patterns can influence cardiac physiology, pathology, and therapies for cardiovascular diseases. Understanding heart development is crucial to evaluating therapeutic eligibility, safety, and efficacy.


Subject(s)
Action Potentials , Adaptation, Physiological , Animals, Newborn , Animals , Guinea Pigs , Age Factors , Heart Rate/physiology , Electrocardiography , Aging/physiology , Isolated Heart Preparation , Calcium Signaling , Male , Heart/physiology , Voltage-Sensitive Dye Imaging , Time Factors , Body Weight , Heart Conduction System/physiology , Female
2.
Biomolecules ; 14(7)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062545

ABSTRACT

Cell-to-cell communication is fundamental to the organization and functionality of multicellular organisms. Intercellular signals orchestrate a variety of cellular responses, including gene expression and protein function changes, and contribute to the integrated functions of individual tissues. Dictyostelium discoideum is a model organism for cell-to-cell interactions mediated by chemical signals and multicellular formation mechanisms. Upon starvation, D. discoideum cells exhibit coordinated cell aggregation via cyclic adenosine 3',5'-monophosphate (cAMP) gradients and chemotaxis, which facilitates the unicellular-to-multicellular transition. During this process, the calcium signaling synchronizes with the cAMP signaling. The resulting multicellular body exhibits organized collective migration and ultimately forms a fruiting body. Various signaling molecules, such as ion signals, regulate the spatiotemporal differentiation patterns within multicellular bodies. Understanding cell-to-cell and ion signaling in Dictyostelium provides insight into general multicellular formation and differentiation processes. Exploring cell-to-cell and ion signaling enhances our understanding of the fundamental biological processes related to cell communication, coordination, and differentiation, with wide-ranging implications for developmental biology, evolutionary biology, biomedical research, and synthetic biology. In this review, I discuss the role of ion signaling in cell motility and development in D. discoideum.


Subject(s)
Cell Movement , Cyclic AMP , Dictyostelium , Signal Transduction , Dictyostelium/metabolism , Dictyostelium/growth & development , Dictyostelium/genetics , Dictyostelium/cytology , Cyclic AMP/metabolism , Chemotaxis , Cell Communication , Ions/metabolism , Cell Differentiation , Calcium Signaling
3.
Biomolecules ; 14(7)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39062578

ABSTRACT

Calcium regulation is essential in virtually any cell due to its critical role as a second messenger in multiple signaling pathways [...].


Subject(s)
Calcium , Drug Discovery , Heart Diseases , Humans , Calcium/metabolism , Drug Discovery/methods , Heart Diseases/metabolism , Heart Diseases/drug therapy , Kinetics , Animals , Calcium Signaling
4.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062821

ABSTRACT

Sensing the lowering of endoplasmic reticulum (ER) calcium (Ca2+), STIM1 mediates a ubiquitous Ca2+ influx process called the store-operated Ca2+ entry (SOCE). Dysregulated STIM1 function or abnormal SOCE is strongly associated with autoimmune disorders, atherosclerosis, and various forms of cancers. Therefore, uncovering the molecular intricacies of post-translational modifications, such as oxidation, on STIM1 function is of paramount importance. In a recent proteomic screening, we identified three protein disulfide isomerases (PDIs)-Prolyl 4-hydroxylase subunit beta (P4HB), protein disulfide-isomerase A3 (PDIA3), and thioredoxin domain-containing protein 5 (TXNDC5)-as the ER-luminal interactors of STIM1. Here, we demonstrated that these PDIs dynamically associate with STIM1 and STIM2. The mutation of the two conserved cysteine residues of STIM1 (STIM1-2CA) decreased its Ca2+ affinity both in cellulo and in situ. Knockdown of PDIA3 or P4HB increased the Ca2+ affinity of wild-type STIM1 while showing no impact on the STIM1-2CA mutant, indicating that PDIA3 and P4HB regulate STIM1's Ca2+ affinity by acting on ER-luminal cysteine residues. This modulation of STIM1's Ca2+ sensitivity was further confirmed by Ca2+ imaging experiments, which showed that knockdown of these two PDIs does not affect STIM1-mediated SOCE upon full store depletion but leads to enhanced SOCE amplitudes upon partial store depletion. Thus, P4HB and PDIA3 dynamically modulate STIM1 activation by fine-tuning its Ca2+ binding affinity, adjusting the level of activated STIM1 in response to physiological cues. The coordination between STIM1-mediated Ca2+ signaling and redox responses reported herein may have implications for cell physiology and pathology.


Subject(s)
Calcium , Neoplasm Proteins , Oxidation-Reduction , Procollagen-Proline Dioxygenase , Protein Disulfide-Isomerases , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 1/genetics , Humans , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/genetics , Calcium/metabolism , Procollagen-Proline Dioxygenase/metabolism , Procollagen-Proline Dioxygenase/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Endoplasmic Reticulum/metabolism , HEK293 Cells , Protein Binding , Calcium Signaling , Stromal Interaction Molecule 2/metabolism , Stromal Interaction Molecule 2/genetics
5.
Int J Mol Sci ; 25(14)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39063213

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have emerged as a promising tool for studying cardiac physiology and drug responses. However, their use is largely limited by an immature phenotype and lack of high-throughput analytical methodology. In this study, we developed a high-throughput testing platform utilizing hPSC-CMs to assess the cardiotoxicity and effectiveness of drugs. Following an optimized differentiation and maturation protocol, hPSC-CMs exhibited mature CM morphology, phenotype, and functionality, making them suitable for drug testing applications. We monitored intracellular calcium dynamics using calcium imaging techniques to measure spontaneous calcium oscillations in hPSC-CMs in the presence or absence of test compounds. For the cardiotoxicity test, hPSC-CMs were treated with various compounds, and calcium flux was measured to evaluate their effects on calcium dynamics. We found that cardiotoxic drugs withdrawn due to adverse drug reactions, including encainide, mibefradil, and cetirizine, exhibited toxicity in hPSC-CMs but not in HEK293-hERG cells. Additionally, in the effectiveness test, hPSC-CMs were exposed to ATX-II, a sodium current inducer for mimicking long QT syndrome type 3, followed by exposure to test compounds. The observed changes in calcium dynamics following drug exposure demonstrated the utility of hPSC-CMs as a versatile model system for assessing both cardiotoxicity and drug efficacy. Overall, our findings highlight the potential of hPSC-CMs in advancing drug discovery and development, which offer a physiologically relevant platform for the preclinical screening of novel therapeutics.


Subject(s)
Cell Differentiation , Drug Evaluation, Preclinical , Myocytes, Cardiac , Pluripotent Stem Cells , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Drug Evaluation, Preclinical/methods , Cell Differentiation/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cardiovascular Agents/pharmacology , Calcium/metabolism , Cardiotoxicity , High-Throughput Screening Assays/methods , HEK293 Cells , Calcium Signaling/drug effects
6.
Front Endocrinol (Lausanne) ; 15: 1412411, 2024.
Article in English | MEDLINE | ID: mdl-39015185

ABSTRACT

Early in the development of Type 2 diabetes (T2D), metabolic stress brought on by insulin resistance and nutrient overload causes ß-cell hyperstimulation. Herein we summarize recent studies that have explored the premise that an increase in the intracellular Ca2+ concentration ([Ca2+]i), brought on by persistent metabolic stimulation of ß-cells, causes ß-cell dysfunction and failure by adversely affecting ß-cell function, structure, and identity. This mini-review builds on several recent reviews that also describe how excess [Ca2+]i impairs ß-cell function.


Subject(s)
Calcium Signaling , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Stress, Physiological , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Humans , Calcium Signaling/physiology , Animals , Stress, Physiological/physiology , Diabetes Mellitus, Type 2/metabolism , Calcium/metabolism , Insulin Resistance/physiology
7.
J Mol Cell Cardiol ; 193: 113-124, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960316

ABSTRACT

The sarcolemmal Ca2+ efflux pathways, Na+-Ca2+-exchanger (NCX) and Ca2+-ATPase (PMCA), play a crucial role in the regulation of intracellular Ca2+ load and Ca2+ transient in cardiomyocytes. The distribution of these pathways between the t-tubular and surface membrane of ventricular cardiomyocytes varies between species and is not clear in human. Moreover, several studies suggest that this distribution changes during the development and heart diseases. However, the consequences of NCX and PMCA redistribution in human ventricular cardiomyocytes have not yet been elucidated. In this study, we aimed to address this point by using a mathematical model of the human ventricular myocyte incorporating t-tubules, dyadic spaces, and subsarcolemmal spaces. Effects of various combinations of t-tubular fractions of NCX and PMCA were explored, using values between 0.2 and 1 as reported in animal experiments under normal and pathological conditions. Small variations in the action potential duration (≤ 2%), but significant changes in the peak value of cytosolic Ca2+ transient (up to 17%) were observed at stimulation frequencies corresponding to the human heart rate at rest and during activity. The analysis of model results revealed that the changes in Ca2+ transient induced by redistribution of NCX and PMCA were mainly caused by alterations in Ca2+ concentrations in the subsarcolemmal spaces and cytosol during the diastolic phase of the stimulation cycle. The results suggest that redistribution of both transporters between the t-tubular and surface membranes contributes to changes in contractility in human ventricular cardiomyocytes during their development and heart disease and may promote arrhythmogenesis.


Subject(s)
Calcium , Heart Ventricles , Myocytes, Cardiac , Sarcolemma , Sodium-Calcium Exchanger , Humans , Myocytes, Cardiac/metabolism , Calcium/metabolism , Sodium-Calcium Exchanger/metabolism , Heart Ventricles/metabolism , Sarcolemma/metabolism , Action Potentials , Calcium Signaling , Cell Membrane/metabolism , Models, Biological , Models, Cardiovascular
8.
Nat Commun ; 15(1): 6131, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033133

ABSTRACT

One question in lymphocyte homing is how integrins are rapidly activated to enable immediate arrest of fast rolling lymphocytes upon encountering chemokines at target vascular beds given the slow chemokine-induced integrin inside-out activation. Herein we demonstrate that chemokine CCL25-triggered Ca2+ influx induces T cell membrane-proximal external Ca2+ concentration ([Ca2+]ex) drop in 6 s from physiological concentration 1.2 mM to 0.3 mM, a critical extracellular Ca2+ threshold for inducing αLß2 activation, triggering rapid αLß2 activation and T cell arrest before occurrence of αLß2 inside-out activation. Talin knockdown inhibits the slow inside-out activation of αLß2 but not [Ca2+]ex drop-triggered αLß2 quick activation. Blocking Ca2+ influx significantly suppresses T cell rolling-to-arrest transition and homing to skin lesions in a mouse psoriasis model, thus alleviating skin inflammation. [Ca2+]ex decrease-triggered rapid integrin activation bridges the gap between initial chemokine stimulation and slow integrin inside-out activation, ensuring immediate lymphocyte arrest and subsequent diapedesis on the right location.


Subject(s)
Calcium , T-Lymphocytes , Talin , Animals , Calcium/metabolism , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Talin/metabolism , Humans , Psoriasis/metabolism , Psoriasis/immunology , Mice, Inbred C57BL , Cell Membrane/metabolism , Integrins/metabolism , Calcium Signaling , Skin/metabolism
9.
Nat Commun ; 15(1): 6143, 2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39034309

ABSTRACT

Wolfram syndrome is a rare genetic disease caused by mutations in the WFS1 or CISD2 gene. A primary defect in Wolfram syndrome involves poor ER Ca2+ handling, but how this disturbance leads to the disease is not known. The current study, performed in primary neurons, the most affected and disease-relevant cells, involving both Wolfram syndrome genes, explains how the disturbed ER Ca2+ handling compromises mitochondrial function and affects neuronal health. Loss of ER Ca2+ content and impaired ER-mitochondrial contact sites in the WFS1- or CISD2-deficient neurons is associated with lower IP3R-mediated Ca2+ transfer from ER to mitochondria and decreased mitochondrial Ca2+ uptake. In turn, reduced mitochondrial Ca2+ content inhibits mitochondrial ATP production leading to an increased NADH/NAD+ ratio. The resulting bioenergetic deficit and reductive stress compromise the health of the neurons. Our work also identifies pharmacological targets and compounds that restore Ca2+ homeostasis, enhance mitochondrial function and improve neuronal health.


Subject(s)
Calcium , Endoplasmic Reticulum , Membrane Proteins , Mitochondria , Neurons , Wolfram Syndrome , Wolfram Syndrome/metabolism , Wolfram Syndrome/genetics , Calcium/metabolism , Mitochondria/metabolism , Endoplasmic Reticulum/metabolism , Animals , Neurons/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Humans , Adenosine Triphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice, Knockout , NAD/metabolism , Calcium Signaling
10.
FASEB J ; 38(14): e23825, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39031532

ABSTRACT

Limb-Girdle Muscular Dystrophy R1/2A (LGMD R1/2A) is caused by mutations in the CAPN3 gene encoding Calpain 3, a skeletal-muscle specific, Ca2+-dependent protease. Localization of Calpain 3 within the triad suggests it contributes to Ca2+ homeostasis. Through live-cell Ca2+ measurements, muscle mechanics, immunofluorescence, and electron microscopy (EM) in Capn3 deficient (C3KO) and wild-type (WT) mice, we determined whether loss of Calpain 3 altered Store-Operated Calcium Entry (SOCE) activity. Direct Ca2+ influx measurements revealed loss of Capn3 elicits elevated resting SOCE and increased resting cytosolic Ca2+, supported by high incidence of calcium entry units (CEUs) observed by EM. C3KO and WT mice were subjected to a single bout of treadmill running to elicit SOCE. Within 1HR post-treadmill running, C3KO mice exhibited diminished force production in extensor digitorum longus muscles and a greater decay of Ca2+ transients in flexor digitorum brevis muscle fibers during repetitive stimulation. Striking evidence for impaired exercise-induced SOCE activation in C3KO mice included poor colocalization of key SOCE proteins, stromal-interacting molecule 1 (STIM1) and ORAI1, combined with disappearance of CEUs in C3KO muscles. These results demonstrate that Calpain 3 is a key regulator of SOCE in skeletal muscle and identify SOCE dysregulation as a contributing factor to LGMD R1/2A pathology.


Subject(s)
Calcium , Calpain , Mice, Knockout , Muscle Proteins , Muscle, Skeletal , Physical Conditioning, Animal , Animals , Calpain/metabolism , Mice , Calcium/metabolism , Muscle Proteins/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Male , Mice, Inbred C57BL , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Calcium Signaling
11.
Sci Signal ; 17(846): eadh2381, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042726

ABSTRACT

The balance of proinflammatory T helper type 17 (TH17) and anti-inflammatory T regulatory (Treg) cells is crucial for immune homeostasis in health and disease. The differentiation of naïve CD4+ T cells into TH17 and Treg cells depends on T cell receptor (TCR) signaling mediated, in part, by interleukin-2-inducible T cell kinase (ITK), which stimulates mitogen-activated protein kinases (MAPKs) and Ca2+ signaling. Here, we report that, in the absence of ITK activity, naïve murine CD4+ T cells cultured under TH17-inducing conditions expressed the Treg transcription factor Foxp3 and did not develop into TH17 cells. Furthermore, ITK inhibition in vivo during allergic inflammation increased the Treg:TH17 ratio in the lung. These switched Foxp3+ Treg-like cells had suppressive function, and their transcriptomic profile resembled that of differentiated, induced Treg (iTreg) cells, but their chromatin accessibility profiles were intermediate between TH17 and iTreg cells. Like iTreg cells, switched Foxp3+ Treg-like cells had reductions in the expression of genes involved in mitochondrial oxidative phosphorylation and glycolysis, in the activation of the mechanistic target of rapamycin (mTOR) signaling pathway, and in the abundance of the TH17 pioneer transcription factor BATF. This ITK-dependent switch between TH17 and Treg cells depended on Ca2+ signaling but not on MAPKs. These findings suggest potential strategies for fine-tuning TCR signal strength through ITK to control the balance of TH17 and Treg cells.


Subject(s)
Cell Differentiation , Forkhead Transcription Factors , Protein-Tyrosine Kinases , T-Lymphocytes, Regulatory , Th17 Cells , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/cytology , Th17 Cells/metabolism , Mice , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Calcium Signaling , Mice, Inbred C57BL , Calcium/metabolism , Mice, Knockout , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/immunology
12.
Planta ; 260(2): 39, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951320

ABSTRACT

MAIN CONCLUSION: Nitrogen stress altered important lipid parameters and related genes in Chlorella pyrenoidosa via ROS and Ca2+ signaling. The mutual interference between ROS and Ca2+ signaling was also uncovered. The changed mechanisms of lipid parameters (especially lipid classes and unsaturation of fatty acids) in microalgae are not completely well known under nitrogen stress. Therefore, Chlorella pyrenoidosa was exposed to 0, 0.5, 1 and 1.5 g L-1 NaNO3 for 4 days. Then, the physiological and biochemical changes were measured. It was shown that the total lipid contents, neutral lipid ratios as well as their related genes (accD and DGAT) increased obviously while the polar lipid ratios, degrees of unsaturation as well as their related genes (PGP and desC) decreased significantly in nitrogen stress groups. The obvious correlations supported that gene expressions should be the necessary pathways to regulate the lipid changes in C. pyrenoidosa under nitrogen stress. The changes in ROS and Ca2+ signaling as well as their significant correlations with corresponding genes and lipid parameters were analyzed. The results suggested that ROS and Ca2+ may regulate these gene expressions and lipid changes in C. pyrenoidosa under nitrogen stress conditions. This was verified by the subordinate tests with an ROS inhibitor and calcium reagents. It also uncovered the clues of mutual interference between ROS and Ca2+ signaling. To summarize, this study revealed the signaling pathways of important lipid changes in microalgae under N stress.


Subject(s)
Chlorella , Nitrogen , Reactive Oxygen Species , Stress, Physiological , Chlorella/metabolism , Chlorella/genetics , Chlorella/physiology , Reactive Oxygen Species/metabolism , Nitrogen/metabolism , Lipid Metabolism/genetics , Calcium/metabolism , Lipids , Calcium Signaling , Signal Transduction , Microalgae/metabolism , Microalgae/genetics
13.
Proc Natl Acad Sci U S A ; 121(29): e2405231121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38990952

ABSTRACT

We report that ~1.8% of all mesothelioma patients and 4.9% of those younger than 55, carry rare germline variants of the BRCA1 associated RING domain 1 (BARD1) gene that were predicted to be damaging by computational analyses. We conducted functional assays, essential for accurate interpretation of missense variants, in primary fibroblasts that we established in tissue culture from a patient carrying the heterozygous BARD1V523A mutation. We found that these cells had genomic instability, reduced DNA repair, and impaired apoptosis. Investigating the underlying signaling pathways, we found that BARD1 forms a trimeric protein complex with p53 and SERCA2 that regulates calcium signaling and apoptosis. We validated these findings in BARD1-silenced primary human mesothelial cells exposed to asbestos. Our study elucidated mechanisms of BARD1 activity and revealed that heterozygous germline BARD1 mutations favor the development of mesothelioma and increase the susceptibility to asbestos carcinogenesis. These mesotheliomas are significantly less aggressive compared to mesotheliomas in asbestos workers.


Subject(s)
Calcium Signaling , DNA Repair , Genetic Predisposition to Disease , Germ-Line Mutation , Mesothelioma , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Humans , DNA Repair/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Mesothelioma/genetics , Calcium Signaling/genetics , Female , Male , Middle Aged , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Fibroblasts/metabolism , Asbestos/toxicity , Genomic Instability
14.
Nat Commun ; 15(1): 5521, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951553

ABSTRACT

The microgeometry of the cellular microenvironment profoundly impacts cellular behaviors, yet the link between it and the ubiquitously expressed mechanosensitive ion channel PIEZO1 remains unclear. Herein, we describe a fluorescent micropipette aspiration assay that allows for simultaneous visualization of intracellular calcium dynamics and cytoskeletal architecture in real-time, under varied micropipette geometries. By integrating elastic shell finite element analysis with fluorescent lifetime imaging microscopy and employing PIEZO1-specific transgenic red blood cells and HEK cell lines, we demonstrate a direct correlation between the microscale geometry of aspiration and PIEZO1-mediated calcium signaling. We reveal that increased micropipette tip angles and physical constrictions lead to a significant reorganization of F-actin, accumulation at the aspirated cell neck, and subsequently amplify the tension stress at the dome of the cell to induce more PIEZO1's activity. Disruption of the F-actin network or inhibition of its mobility leads to a notable decline in PIEZO1 mediated calcium influx, underscoring its critical role in cellular mechanosensing amidst geometrical constraints.


Subject(s)
Actins , Calcium , Cytoskeleton , Ion Channels , Mechanotransduction, Cellular , Humans , Ion Channels/metabolism , Actins/metabolism , HEK293 Cells , Cytoskeleton/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Finite Element Analysis , Animals , Microscopy, Fluorescence/methods
15.
Methods Mol Biol ; 2816: 69-75, 2024.
Article in English | MEDLINE | ID: mdl-38977589

ABSTRACT

Intracellular Ca2+ can be conveniently monitored by sensitive Ca2+ fluorescent dyes in live cells. The Gαq involved lipid signaling pathways and, thus, can be studied by intracellular Ca2+ imaging. Here we describe the protocols to measure intracellular Ca2+ for studying PEG2-EP1 activity in esophageal smooth muscle cells. The ratiometric Fura-2 imaging provides quantitative data, and the Fluo-4 confocal microscopic imaging has high-spatial resolution.


Subject(s)
Calcium , Receptors, G-Protein-Coupled , Calcium/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Microscopy, Confocal/methods , Signal Transduction , Myocytes, Smooth Muscle/metabolism , Calcium Signaling , Humans , Xanthenes/metabolism , Fura-2/metabolism , Lipid Metabolism , Esophagus/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Aniline Compounds
19.
FASEB J ; 38(13): e23737, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38953724

ABSTRACT

Meningiomas are the most common primary intracranial tumors and account for nearly 30% of all nervous system tumors. Approximately half of meningioma patients exhibit neurofibromin 2 (NF2) gene inactivation. Here, NF2 was shown to interact with the endoplasmic reticulum (ER) calcium (Ca2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in IOMM-Lee, a high-grade malignant meningioma cell line, and the F1 subdomain of NF2 plays a critical role in this interaction. Functional assays indicated that NF2 promotes the phosphorylation of IP3R (Ser 1756) and IP3R-mediated endoplasmic reticulum (ER) Ca2+ release by binding to IP3R1, which results in Ca2+-dependent apoptosis. Knockout of NF2 decreased Ca2+ release and promoted resistance to apoptosis, which was rescued by wild-type NF2 overexpression but not by F1 subdomain deletion truncation overexpression. The effects of NF2 defects on the development of tumors were further studied in mouse models. The decreased expression level of NF2 caused by NF2 gene knockout or mutation affects the activity of the IP3R channel, which reduces Ca2+-dependent apoptosis, thereby promoting the development of tumors. We elucidated the interaction patterns of NF2 and IP3R1, revealed the molecular mechanism through which NF2 regulates IP3R1-mediated Ca2+ release, and elucidated the new pathogenic mechanism of meningioma-related NF2 variants. Our study broadens the current understanding of the biological function of NF2 and provides ideas for drug screening of NF2-associated meningioma.


Subject(s)
Apoptosis , Calcium Signaling , Calcium , Inositol 1,4,5-Trisphosphate Receptors , Meningeal Neoplasms , Meningioma , Animals , Humans , Mice , Calcium/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningeal Neoplasms/genetics , Meningioma/metabolism , Meningioma/pathology , Meningioma/genetics , Neurofibromin 2
20.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000053

ABSTRACT

Sclerotinia sclerotiorum (Ss) is one of the most devastating fungal pathogens, causing huge yield loss in multiple economically important crops including oilseed rape. Plant resistance to Ss pertains to quantitative disease resistance (QDR) controlled by multiple minor genes. Genome-wide identification of genes involved in QDR to Ss is yet to be conducted. In this study, we integrated several assays including genome-wide association study (GWAS), multi-omics co-localization, and machine learning prediction to identify, on a genome-wide scale, genes involved in the oilseed rape QDR to Ss. Employing GWAS and multi-omics co-localization, we identified seven resistance-associated loci (RALs) associated with oilseed rape resistance to Ss. Furthermore, we developed a machine learning algorithm and named it Integrative Multi-Omics Analysis and Machine Learning for Target Gene Prediction (iMAP), which integrates multi-omics data to rapidly predict disease resistance-related genes within a broad chromosomal region. Through iMAP based on the identified RALs, we revealed multiple calcium signaling genes related to the QDR to Ss. Population-level analysis of selective sweeps and haplotypes of variants confirmed the positive selection of the predicted calcium signaling genes during evolution. Overall, this study has developed an algorithm that integrates multi-omics data and machine learning methods, providing a powerful tool for predicting target genes associated with specific traits. Furthermore, it makes a basis for further understanding the role and mechanisms of calcium signaling genes in the QDR to Ss.


Subject(s)
Ascomycota , Brassica napus , Calcium Signaling , Disease Resistance , Genome-Wide Association Study , Machine Learning , Plant Diseases , Ascomycota/pathogenicity , Disease Resistance/genetics , Plant Diseases/microbiology , Plant Diseases/genetics , Brassica napus/genetics , Brassica napus/microbiology , Brassica napus/immunology , Calcium Signaling/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Genomics/methods , Multiomics
SELECTION OF CITATIONS
SEARCH DETAIL