943 results on '"Cav1.2"'
Search Results
2. Altered Protein Kinase A-Dependent Phosphorylation of Cav1.2 in Left Ventricular Myocardium from Cacna1c Haploinsufficient Rat Hearts.
- Author
-
Königstein, David, Fender, Hauke, Plačkić, Jelena, Kisko, Theresa M., Wöhr, Markus, and Kockskämper, Jens
- Abstract
CACNA1C encodes the α1c subunit of the L-type Ca2+ channel, Cav1.2. Ventricular myocytes from haploinsufficient Cacna1c (Cacna1c+/−) rats exhibited reduced expression of Cav1.2 but an apparently normal sarcolemmal Ca2+ influx with an impaired response to sympathetic stress. We tested the hypothesis that the altered phosphorylation of Cav1.2 might underlie the sarcolemmal Ca2+ influx phenotype in Cacna1c+/− myocytes using immunoblotting of the left ventricular (LV) tissue from Cacna1c+/− versus wildtype (WT) hearts. Activation of cAMP-dependent protein kinase A (PKA) increases L-type Ca2+ current and phosphorylates Cav1.2 at serine-1928. Using an antibody directed against this phosphorylation site, we observed elevated phosphorylation of Cav1.2 at serine-1928 in LV myocardium from Cacna1c+/− rats under basal conditions (+110% versus WT). Sympathetic stress was simulated by isoprenaline (100 nM) in Langendorff-perfused hearts. Isoprenaline increased the phosphorylation of serine-1928 in Cacna1c+/− LV myocardium by ≈410%, but the increase was significantly smaller than in WT myocardium (≈650%). In conclusion, our study reveals altered PKA-dependent phosphorylation of Cav1.2 with elevated phosphorylation of serine-1928 under basal conditions and a diminished phosphorylation reserve during β-adrenergic stimulation. These alterations in the phosphorylation of Cav1.2 may explain the apparently normal sarcolemmal Ca2+ influx in Cacna1c+/− myocytes under basal conditions as well as the impaired response to sympathetic stimulation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Tetrodotoxin‐resistant mechanosensitivity and L‐type calcium channel‐mediated spontaneous calcium activity in enteric neurons
- Author
-
Richard J. Amedzrovi Agbesi, Amira El Merhie, Nick J. Spencer, Tim Hibberd, and Nicolas R. Chevalier
- Subjects
2‐APB ,adult gut ,calcium imaging ,CaV1.2 ,embryonic gut ,enteric nervous system ,Physiology ,QP1-981 - Abstract
Abstract Gut motility undergoes a switch from myogenic to neurogenic control in late embryonic development. Here, we report on the electrical events that underlie this transition in the enteric nervous system, using the GCaMP6f reporter in neural crest cell derivatives. We found that spontaneous calcium activity is tetrodotoxin (TTX) resistant at stage E11.5, but not at E18.5. Motility at E18.5 was characterized by periodic, alternating high‐ and low‐frequency contractions of the circular smooth muscle; this frequency modulation was inhibited by TTX. Calcium imaging at the neurogenic‐motility stages E18.5–P3 showed that CaV1.2‐positive neurons exhibited spontaneous calcium activity, which was inhibited by nicardipine and 2‐aminoethoxydiphenyl borate (2‐APB). Our protocol locally prevented muscle tone relaxation, arguing for a direct effect of nicardipine on enteric neurons, rather than indirectly by its relaxing effect on muscle. We demonstrated that the ENS was mechanosensitive from early stages on (E14.5) and that this behaviour was TTX and 2‐APB resistant. We extended our results on L‐type channel‐dependent spontaneous activity and TTX‐resistant mechanosensitivity to the adult colon. Our results shed light on the critical transition from myogenic to neurogenic motility in the developing gut, as well as on the intriguing pathways mediating electro‐mechanical sensitivity in the enteric nervous system. Highlights What is the central question of this study? What are the first neural electric events underlying the transition from myogenic to neurogenic motility in the developing gut, what channels do they depend on, and does the enteric nervous system already exhibit mechanosensitivity? What is the main finding and its importance? ENS calcium activity is sensitive to tetrodotoxin at stage E18.5 but not E11.5. Spontaneous electric activity at fetal and adult stages is crucially dependent on L‐type calcium channels and IP3R receptors, and the enteric nervous system exhibits a tetrodotoxin‐resistant mechanosensitive response. Abstract figure legend Tetrodotoxin‐resistant Ca2+ rise induced by mechanical stimulation in the E18.5 mouse duodenum.
- Published
- 2024
- Full Text
- View/download PDF
4. Enhanced isradipine sensitivity in vascular smooth muscle cells due to hypoxia‐induced Cav1.2 splicing and RbFox1/Fox2 downregulation.
- Author
-
Poore, Charlene Priscilla, Yang, Jialei, Wei, Shunhui, Fhu, Chee Kong, Bichler, Zoë, Wang, Juejin, Soong, Tuck Wah, and Liao, Ping
- Subjects
- *
VASCULAR smooth muscle , *ALTERNATIVE RNA splicing , *MUSCLE cells , *CALCIUM antagonists , *SMOOTH muscle , *CALCIUM channels - Abstract
Calcium influx via the L‐type voltage‐gated Cav1.2 calcium channel in smooth muscle cells regulates vascular contraction. Calcium channel blockers (CCBs) are widely used to treat hypertension by inhibiting Cav1.2 channels. Using the vascular smooth muscle cell line, A7r5 and primary culture of cerebral vascular smooth muscle cells, we found that the expression and function of Cav1.2 channels are downregulated during hypoxia. Furthermore, hypoxia induces structural changes in Cav1.2 channels via alternative splicing. The expression of exon 9* is upregulated, whereas exon 33 is downregulated. Such structural alterations of Cav1.2 channels are caused by the decreased expression of RNA‐binding proteins RNA‐binding protein fox‐1 homolog 1 and 2 (RbFox1 and RbFox2). Overexpression of RbFox1 and RbFox2 prevents hypoxia‐induced exon 9* inclusion and exon 33 exclusion. Importantly, such structural alterations of the Cav1.2 channel partly contribute to the enhanced sensitivity of Cav1.2 to isradipine (a CCB) under hypoxia. Overexpression of RbFox1 and RbFox2 successfully reduces isradipine sensitivity in hypoxic smooth muscle cells. Our results suggest a new strategy to manage ischemic diseases such as stroke and myocardial infarction. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Tetrodotoxin‐resistant mechanosensitivity and L‐type calcium channel‐mediated spontaneous calcium activity in enteric neurons.
- Author
-
Amedzrovi Agbesi, Richard J., El Merhie, Amira, Spencer, Nick J., Hibberd, Tim, and Chevalier, Nicolas R.
- Subjects
- *
ENTERIC nervous system , *SMOOTH muscle contraction , *CALCIUM channels , *NEURAL crest , *MUSCLE tone - Abstract
Gut motility undergoes a switch from myogenic to neurogenic control in late embryonic development. Here, we report on the electrical events that underlie this transition in the enteric nervous system, using the GCaMP6f reporter in neural crest cell derivatives. We found that spontaneous calcium activity is tetrodotoxin (TTX) resistant at stage E11.5, but not at E18.5. Motility at E18.5 was characterized by periodic, alternating high‐ and low‐frequency contractions of the circular smooth muscle; this frequency modulation was inhibited by TTX. Calcium imaging at the neurogenic‐motility stages E18.5–P3 showed that CaV1.2‐positive neurons exhibited spontaneous calcium activity, which was inhibited by nicardipine and 2‐aminoethoxydiphenyl borate (2‐APB). Our protocol locally prevented muscle tone relaxation, arguing for a direct effect of nicardipine on enteric neurons, rather than indirectly by its relaxing effect on muscle. We demonstrated that the ENS was mechanosensitive from early stages on (E14.5) and that this behaviour was TTX and 2‐APB resistant. We extended our results on L‐type channel‐dependent spontaneous activity and TTX‐resistant mechanosensitivity to the adult colon. Our results shed light on the critical transition from myogenic to neurogenic motility in the developing gut, as well as on the intriguing pathways mediating electro‐mechanical sensitivity in the enteric nervous system. Highlights: What is the central question of this study?What are the first neural electric events underlying the transition from myogenic to neurogenic motility in the developing gut, what channels do they depend on, and does the enteric nervous system already exhibit mechanosensitivity?What is the main finding and its importance?ENS calcium activity is sensitive to tetrodotoxin at stage E18.5 but not E11.5. Spontaneous electric activity at fetal and adult stages is crucially dependent on L‐type calcium channels and IP3R receptors, and the enteric nervous system exhibits a tetrodotoxin‐resistant mechanosensitive response. Abstract figure legend Tetrodotoxin‐resistant Ca2+ rise induced by mechanical stimulation in the E18.5 mouse duodenum. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Mechanism of gabapentinoid potentiation of opioid effects on cyclic AMP signaling in neuropathic pain.
- Author
-
Garza-Carbajal, Anibal, Bavencoffe, Alexis, Herrera, Juan J., Johnson, Kayla N., Walters, Edgar T., and Dessauer, Carmen W.
- Subjects
- *
DORSAL root ganglia , *CYCLIC adenylic acid , *ADENYLATE cyclase , *CALCIUM channels , *SPINAL cord injuries - Abstract
Over half of spinal cord injury (SCI) patients develop opioid-resistant chronic neuropathic pain. Safer alternatives to opioids for treatment of neuropathic pain are gabapentinoids (e.g., pregabalin and gabapentin). Clinically, gabapentinoids appear to amplify opioid effects, increasing analgesia and overdose-related adverse outcomes, but in vitro proof of this amplification and its mechanism are lacking. We previously showed that after SCI, sensitivity to opioids is reduced by fourfold to sixfold in rat sensory neurons. Here, we demonstrate that after injury, gabapentinoids restore normal sensitivity of opioid inhibition of cyclic AMP (cAMP) generation, while reducing nociceptor hyperexcitability by inhibiting voltage-gated calcium channels (VGCCs). Increasing intracellular Ca2+ or activation of L-type VGCCs (L-VGCCs) suffices to mimic SCI effects on opioid sensitivity, in a manner dependent on the activity of the Raf1 proto-oncogene, serine/threonine-protein kinase C-Raf, but independent of neuronal depolarization. Together, our results provide a mechanism for potentiation of opioid effects by gabapentinoids after injury, via reduction of calcium influx through L-VGCCs, and suggest that other inhibitors targeting these channels may similarly enhance opioid treatment of neuropathic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Evaluation of four KCNMA1 channelopathy variants on BK channel current under CaV1.2 activation
- Author
-
Ria L. Dinsdale and Andrea L. Meredith
- Subjects
KCa1.1 ,calcium-activated potassium channel ,channelopathy ,CaV1.2 ,voltage-gated calcium channels ,CACNA1C ,Therapeutics. Pharmacology ,RM1-950 ,Physiology ,QP1-981 - Abstract
Variants in KCNMA1, encoding the voltage- and calcium-activated K+ (BK) channel, are associated with human neurological disease. The effects of gain-of-function (GOF) and loss-of-function (LOF) variants have been predominantly studied on BK channel currents evoked under steady-state voltage and Ca2+ conditions. However, in their physiological context, BK channels exist in partnership with voltage-gated Ca2+ channels and respond to dynamic changes in intracellular Ca2+ (Ca2+i). In this study, an L-type voltage-gated Ca2+ channel present in the brain, CaV1.2, was co-expressed with wild type and mutant BK channels containing GOF (D434G, N999S) and LOF (H444Q, D965V) patient-associated variants in HEK-293T cells. Whole-cell BK currents were recorded under CaV1.2 activation using buffering conditions that restrict Ca2+i to nano- or micro-domains. Both conditions permitted wild type BK current activation in response to CaV1.2 Ca2+ influx, but differences in behavior between wild type and mutant BK channels were reduced compared to prior studies in clamped Ca2+i. Only the N999S mutation produced an increase in BK current in both micro- and nano-domains using square voltage commands and was also detectable in BK current evoked by a neuronal action potential within a microdomain. These data corroborate the GOF effect of N999S on BK channel activity under dynamic voltage and Ca2+ stimuli, consistent with its pathogenicity in neurological disease. However, the patient-associated mutations D434G, H444Q, and D965V did not exhibit significant effects on BK current under CaV1.2-mediated Ca2+ influx, in contrast with prior steady-state protocols. These results demonstrate a differential potential for KCNMA1 variant pathogenicity compared under diverse voltage and Ca2+ conditions.
- Published
- 2024
- Full Text
- View/download PDF
8. Depression of CaV1.2 activation and expression in mast cells ameliorates allergic inflammation diseases
- Author
-
Yongjing Zhang, Yingnan Zeng, Haoyun Bai, Wen Zhang, Zhuoyin Xue, Shiling Hu, Shemin Lu, and Nan Wang
- Subjects
Allergy ,Asthma ,Mast cell ,CaV1.2 ,CaMKII/PKC ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Allergic inflammation is closely related to the activation of mast cells (MCs), which is regulated by its intracellular Ca2+ level, but the intake and effects of the intracellular Ca2+ remain unclear. The Ca2+ influx is controlled by members of Ca2+ channels, among which calcium voltage-gated channel subunit alpha1 C (CaV1.2) is the most robust. This study aimed to reveal the role and underlying mechanism of MC CaV1.2 in allergic inflammation. We found that CaV1.2 participated in MC activation and allergic inflammation. Nimodipine (Nim), as a strong CaV1.2-specific antagonist, ameliorated allergic inflammation in mice. Further, CaV1.2 activation in MC was triggered by phosphatizing at its Ser1928 through protein kinase C (PKC), which calcium/calmodulin-dependent protein kinase II (CaMKII) catalyzed. Overexpression or knockdown of MC CaV1.2 influenced MC activation. Importantly, CaV1.2 expression in MC had detrimental effects, while its deficiency ameliorated allergic pulmonary inflammation. Results provide novel insights into CaV1.2 function and a potential drug target for controlling allergic inflammation.
- Published
- 2024
- Full Text
- View/download PDF
9. Eicosapentaenoic Acid Rescues Cav1.2-L-Type Ca 2+ Channel Decline Caused by Saturated Fatty Acids via Both Free Fatty Acid Receptor 4-Dependent and -Independent Pathways in Cardiomyocytes.
- Author
-
Morishima, Masaki, Wang, Pu, Horii, Kosuke, Horikawa, Kazuki, and Ono, Katsushige
- Subjects
- *
SATURATED fatty acids , *CALCIUM ions , *UNSATURATED fatty acids , *PALMITIC acid , *OMEGA-3 fatty acids , *FREE fatty acids , *EICOSAPENTAENOIC acid - Abstract
Dietary intake of omega-3 polyunsaturated fatty acids (eicosapentaenoic acid, EPA) exerts antiarrhythmic effects, although the mechanisms are poorly understood. Here, we investigated the possible beneficial actions of EPA on saturated fatty acid-induced changes in the L-type Ca2+ channel in cardiomyocytes. Cardiomyocytes were cultured with an oleic acid/palmitic acid mixture (OAPA) in the presence or absence of EPA. Beating rate reduction in cardiomyocytes caused by OAPA were reversed by EPA. EPA also retrieved a reduction in Cav1.2 L-type Ca2+ current, mRNA, and protein caused by OAPA. Immunocytochemical analysis revealed a distinct downregulation of the Cav1.2 channel caused by OAPA with a concomitant decrease in the phosphorylated component of a transcription factor adenosine-3′,5′-cyclic monophosphate (cAMP) response element binding protein (CREB) in the nucleus, which were rescued by EPA. A free fatty acid receptor 4 (FFAR4) agonist TUG-891 reversed expression of Cav1.2 and CREB mRNA caused by OAPA, whereas an FFAR4 antagonist AH-7614 abolished the effects of EPA. Excessive reactive oxygen species (ROS) accumulation caused by OAPA decreased Cav1.2 and CREB mRNA expressions, which was reversed by an ROS scavenger. Our data suggest that EPA rescues cellular Cav1.2-Ca2+ channel decline caused by OAPA lipotoxicity and oxidative stresses via both free fatty acid receptor 4-dependent and -independent pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Calcium Channels, Clustering, and Single‐Amino Acid Control of Blood Pressure and Blood Flow
- Author
-
Lars Jørn Jensen
- Subjects
Editorials ,amino acid phosphorylation ,blood flow regulation ,CaV1.2 ,hypertension ,ion channel clustering ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2024
- Full Text
- View/download PDF
11. Calmodulin promotes a Ca2+‐dependent conformational change in the C‐terminal regulatory domain of CaV1.2
- Author
-
Yadav, Deepak Kumar, Anderson, David E, Hell, Johannes W, and Ames, James B
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,2.1 Biological and endogenous factors ,Calmodulin ,Calcium ,Protein Binding ,calmodulin ,Ca(V)1 ,2 ,4 ,IQ-switch ,L-type Ca2+ channel ,NMR ,CaV1.2 ,CaV1.4 ,Medicinal and Biomolecular Chemistry ,Evolutionary Biology ,Biochemistry & Molecular Biology ,Biochemistry and cell biology - Abstract
Calmodulin (CaM) binds to the membrane-proximal cytosolic C-terminal domain of CaV 1.2 (residues 1520-1669, CT(1520-1669)) and causes Ca2+ -induced conformational changes that promote Ca2+ -dependent channel inactivation (CDI). We report biophysical studies that probe the structural interaction between CT(1520-1669) and CaM. The recombinantly expressed CT(1520-1669) is insoluble, but can be solubilized in the presence of Ca2+ -saturated CaM (Ca4 /CaM), but not in the presence of Ca2+ -free CaM (apoCaM). We show that half-calcified CaM (Ca2 /CaM12 ) forms a complex with CT(1520-1669) that is less soluble than CT(1520-1669) bound to Ca4 /CaM. The NMR spectrum of CT(1520-1669) reveals spectral differences caused by the binding of Ca2 /CaM12 versus Ca4 /CaM, suggesting that the binding of Ca2+ to the CaM N-lobe may induce a conformational change in CT(1520-1669).
- Published
- 2022
12. Chemical shift assignments of the C-terminal domain of CaBP1 bound to the IQ-motif of voltage-gated Ca2+ channel (CaV1.2)
- Author
-
Salveson, Ian and Ames, James B
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Calcium ,Calcium-Binding Proteins ,Calmodulin ,Nuclear Magnetic Resonance ,Biomolecular ,CaBP1 ,EF-hand ,Ca(V)1 ,2 ,IQ-motif ,CDI ,CaV1.2 ,Biophysics ,Biochemistry and cell biology - Abstract
The neuronal L-type voltage-gated Ca2+ channel (CaV1.2) interacts with Ca2+ binding protein 1 (CaBP1), that promotes Ca2+-induced channel activity. The binding of CaBP1 to the IQ-motif in CaV1.2 (residues 1644-1665) blocks the binding of calmodulin and prevents Ca2+-dependent inactivation of CaV1.2. This Ca2+-induced binding of CaBP1 to CaV1.2 is important for modulating neuronal synaptic plasticity, which may serve a role in learning and memory. Here we report NMR assignments of the C-terminal domain of CaBP1 (residues 99-167, called CaBP1C) that contains two Ca2+ bound at the third and fourth EF-hands (EF3 and EF4) and is bound to the CaV1.2 IQ-motif from CaV1.2 (BMRB accession no. 51518).
- Published
- 2022
13. Ambroxol-Enhanced Frequency and Amplitude of Beating Cilia Controlled by a Voltage-Gated Ca 2+ Channel, Cav1.2, via pH i Increase and [Cl − ] i Decrease in the Lung Airway Epithelial Cells of Mice.
- Author
-
Nakahari, Takashi, Suzuki, Chihiro, Kawaguchi, Kotoku, Hosogi, Shigekuni, Tanaka, Saori, Asano, Shinji, Inui, Toshio, and Marunaka, Yoshinori
- Subjects
- *
CALCIUM ions , *EPITHELIAL cells , *CILIA & ciliary motion , *LUNGS , *MICE , *CELL size - Abstract
Ambroxol (ABX), a frequently prescribed secretolytic agent which enhances the ciliary beat frequency (CBF) and ciliary bend angle (CBA, an index of amplitude) by 30%, activates a voltage-dependent Ca2+ channel (CaV1.2) and a small transient Ca2+ release in the ciliated lung airway epithelial cells (c-LAECs) of mice. The activation of CaV1.2 alone enhanced the CBF and CBA by 20%, mediated by a pHi increasei and a [Cl−]i decrease in the c-LAECs. The increase in pHi, which was induced by the activation of the Na+-HCO3− cotransporter (NBC), enhanced the CBF (by 30%) and CBA (by 15–20%), and a decrease in [Cl−]i, which was induced by the Cl− release via anoctamine 1 (ANO1), enhanced the CBA (by 10–15%). While a Ca2+-free solution or nifedipine (an inhibitor of CaV1.2) inhibited 70% of the CBF and CBA enhancement using ABX, CaV1.2 enhanced most of the CBF and CBA increases using ABX. The activation of the CaV1.2 existing in the cilia stimulates the NBC to increase pHi and ANO1 to decrease the [Cl−]i in the c-LAECs. In conclusion, the pHi increase and the [Cl−]i decrease enhanced the CBF and CBA in the ABX-stimulated c-LAECs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. CACNA1C-Related Channelopathies
- Author
-
Herold, Kevin G., Hussey, John W., Dick, Ivy E., Michel, Martin C., Editor-in-Chief, Barrett, James E., Editorial Board Member, Centurión, David, Editorial Board Member, Flockerzi, Veit, Editorial Board Member, Geppetti, Pierangelo, Editorial Board Member, Hofmann, Franz B., Editorial Board Member, Meier, Kathryn Elaine, Editorial Board Member, Page, Clive P., Editorial Board Member, Wang, KeWei, Editorial Board Member, and Striessnig, Jörg, editor
- Published
- 2023
- Full Text
- View/download PDF
15. Fibrillin-1 mutation contributes to Marfan syndrome by inhibiting Cav1.2-mediated cell proliferation in vascular smooth muscle cells
- Author
-
Wenfeng Lin, Jiaqi Xiong, Yefan Jiang, Hao Liu, Jinhui Bian, Juejin Wang, Yongfeng Shao, and Buqing Ni
- Subjects
Marfan Syndrome ,Cav1.2 ,FBN1 ,cell proliferation ,cell cycle ,Therapeutics. Pharmacology ,RM1-950 ,Physiology ,QP1-981 - Abstract
ABSTRACTMarfan syndrome (MFS) is an autosomal dominant connective tissue disorder caused by mutation in fibrillin-1 (FBN1). However, the molecular mechanism underlying MFS remains poorly understood. The study aimed to explore how the L-type calcium channel (CaV1.2) modulates disease progression of MFS and to identify a potential effective target for attenuating MFS. KEGG enrichment analysis showed that the calcium signaling pathway gene set was significantly enriched. We demonstrated that FBN1 deficiency exhibited inhibition on both the expression of Cav1.2 and proliferation of vascular smooth muscle cells (VSMCs). Then, we examined whether FBN1 mediates Cav1.2 via regulating TGF-β1. Higher levels of TGF-β1 were observed in the serum and aortic tissues from patients with MFS. TGF-β1 modulated Cav1.2 expression in a concentration-dependent manner. We evaluated the role of Cav1.2 in MFS by small interfering RNA and Cav1.2 agonist Bay K8644. The effect of Cav1.2 on cell proliferation was dependent on c-Fos activity. These results demonstrated FBN1 deficiency decreased the expression levels of Cav1.2 via regulation of TGF-β1, and downregulation of Cav1.2 inhibited cell proliferation of human aortic smooth muscle cells (HASMCs) in MFS patients. These findings suggest that Cav1.2 may be an appealing therapeutic target for MFS.
- Published
- 2023
- Full Text
- View/download PDF
16. L-type calcium channels and neuropsychiatric diseases: Insights into genetic risk variant-associated genomic regulation and impact on brain development
- Author
-
Madelyn R. Baker, Andrew S. Lee, and Anjali M. Rajadhyaksha
- Subjects
CACNA1C ,CACNA1D ,Cav1.2 ,Cav1.3 ,neuropsychiatric ,Therapeutics. Pharmacology ,RM1-950 ,Physiology ,QP1-981 - Abstract
ABSTRACTRecent human genetic studies have linked a variety of genetic variants in the CACNA1C and CACNA1D genes to neuropsychiatric and neurodevelopmental disorders. This is not surprising given the work from multiple laboratories using cell and animal models that have established that Cav1.2 and Cav1.3 L-type calcium channels (LTCCs), encoded by CACNA1C and CACNA1D, respectively, play a key role in various neuronal processes that are essential for normal brain development, connectivity, and experience-dependent plasticity. Of the multiple genetic aberrations reported, genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) in CACNA1C and CACNA1D that are present within introns, in accordance with the growing body of literature establishing that large numbers of SNPs associated with complex diseases, including neuropsychiatric disorders, are present within non-coding regions. How these intronic SNPs affect gene expression has remained a question. Here, we review recent studies that are beginning to shed light on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic and chromatin levels. We additionally review recent studies that are uncovering how altered calcium signaling through LTCCs impact some of the neuronal developmental processes, such as neurogenesis, neuron migration, and neuron differentiation. Together, the described changes in genomic regulation and disruptions in neurodevelopment provide possible mechanisms by which genetic variants of LTCC genes contribute to neuropsychiatric and neurodevelopmental disorders.
- Published
- 2023
- Full Text
- View/download PDF
17. L-Type Ca2+ Channel Regulation by Calmodulin and CaBP1
- Author
-
Ames, James B
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,1.1 Normal biological development and functioning ,Calmodulin ,Humans ,Animals ,Calcium Channels ,L-Type ,Calcium-Binding Proteins ,Calcium ,Protein Binding ,calmodulin ,CaBP1 ,CaV1 ,2 ,3 ,L-type Ca2+ channel ,EF-hand ,IQ-motif ,CaV1.2 ,CaV1.3 ,Biochemistry and Cell Biology ,Biochemistry and cell biology ,Bioinformatics and computational biology ,Medical biotechnology - Abstract
L-type voltage-gated Ca2+ channels (CaV1.2 and CaV1.3, called CaV) interact with the Ca2+ sensor proteins, calmodulin (CaM) and Ca2+ binding Protein 1 (CaBP1), that oppositely control Ca2+-dependent channel activity. CaM and CaBP1 can each bind to the IQ-motif within the C-terminal cytosolic domain of CaV, which promotes increased channel open probability under basal conditions. At elevated cytosolic Ca2+ levels (caused by CaV channel opening), Ca2+-bound CaM binding to CaV is essential for promoting rapid Ca2+-dependent channel inactivation (CDI). By contrast, CaV binding to CaBP1 prevents CDI and promotes Ca2+-induced channel opening (called CDF). In this review, I provide an overview of the known structures of CaM and CaBP1 and their structural interactions with the IQ-motif to help understand how CaM promotes CDI, whereas CaBP1 prevents CDI and instead promotes CDF. Previous electrophysiology studies suggest that Ca2+-free forms of CaM and CaBP1 may pre-associate with CaV under basal conditions. However, previous Ca2+ binding data suggest that CaM and CaBP1 are both calculated to bind to Ca2+ with an apparent dissociation constant of ~100 nM when CaM or CaBP1 is bound to the IQ-motif. Since the neuronal basal cytosolic Ca2+ concentration is ~100 nM, nearly half of the neuronal CaV channels are suggested to be bound to Ca2+-bound forms of either CaM or CaBP1 under basal conditions. The pre-association of CaV with calcified forms of CaM or CaBP1 are predicted here to have functional implications. The Ca2+-bound form of CaBP1 is proposed to bind to CaV under basal conditions to block CaV binding to CaM, which could explain how CaBP1 might prevent CDI.
- Published
- 2021
18. Wildtype peers rescue social play and 50-kHz ultrasonic vocalization deficits in juvenile female Cacna1c heterozygous rats.
- Author
-
Bogdan, Rebecca, Kayumova, Rukhshona, Schwarting, Rainer K. W., Wöhr, Markus, and Kisko, Theresa M.
- Subjects
FEMALES ,SOUNDS ,PEER pressure ,RATS ,NEUROBEHAVIORAL disorders ,SOCIAL influence ,ANIMAL social behavior - Abstract
Background: Healthy brain development depends on early social practices and experiences. The risk gene CACNA1C is implicated in numerous neuropsychiatric disorders, in which key characteristics include deficits in social functioning and communication. Recently, we reported sex-dependent impairments in social behavior and ultrasonic vocalizations (USV) in juvenile heterozygous Cacna1c
+/- (HET) rats. Specifically, HET females displayed increases in rough-and-tumble play that eliminated the typically observed sex difference between male and female rats. Interestingly, female wild-type Cacna1c+/+ (WT) pairs also showed a similar increase in social play when housed with HET females, suggesting their behavior may be influenced by HET cage mates. This indicates that the genetic makeup of the social environment related to Cacna1c can influence social play, yet systematic studies are lacking. Methods: In the present study, we housed juvenile females in MIXED- or SAME-genotype cages and tested them in a social play paradigm with a same- and opposite-genotype partner. Results: The results show that the early social environment and the genotype of the play partner influence social play and 50-kHz USV emission. Experience with a WT play partner appears necessary for HET females to show comparable levels of play and 50-kHz USV emission. Same-genotype HET pairs played less and emitted fewer 50-kHz USV than same-genotype WT or opposite-genotype pairs; however, we found that the decrease in social play and 50-kHz USV in HET pairs can be rescued by playing with a WT partner. The effect was particularly prominent when the first play partner was WT, as we found it increased play and 50-kHz USV emission in all subsequent interactions with ensuing partners. Conclusion: These findings suggest that the genetic makeup related to the social environment and/or social peers influences social play in Cacna1c+/- haploinsufficient rats. Specifically, our results show that WT peers can rescue behavior and communication alterations in Cacna1c female rats. Our findings have important implications because they show that the genetic makeup of the social environment can divulge phenotypic changes in genetic rat models of neuropsychiatric disorders. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
19. Prenatal dexamethasone exposure impaired vascular reactivity in adult male offspring cerebral arteries.
- Author
-
Lei, Jiahui, Zhao, Meng, Deng, Fengying, Xu, Ting, Ji, Bingyu, Wang, Xietong, Zhang, Meihua, Sun, Miao, and Gao, Qinqin
- Subjects
- *
PRENATAL exposure , *GENE expression , *ANGIOTENSIN II , *CEREBROVASCULAR disease , *SPRAGUE Dawley rats , *CONTRACTILITY (Biology) , *CEREBRAL arteries - Abstract
Cerebrovascular disease is one of the leading causes of death worldwide. Middle cerebral artery (MCA) is the largest and most complex of cerebral arteries. The prenatal period is a critical time for development, which largely determines lifelong health. Clinically, glucocorticoids (GCs) administration to accelerate preterm fetal lung maturation has become standard practice. Prenatal GCs administration increases cardiovascular risks in offspring, but little is known regarding the side effects on offspring MCA function. We investigated the alterations of MCA reactivity following prenatal GCs administration in postnatal offspring. Pregnant Sprague-Dawley rats received synthetic GCs (dexamethasone, DEX) during the last week of pregnancy, and we examined vascular reactivity, cellular electrophysiology, and gene promoter epigenetic modifications in the male offspring MCA. Our results showed that prenatal DEX exposure increased the sensitivity of offspring MCA to Angiotensin II, which was resulted from the increased Cav1.2 (L-type Ca2+ channels subunit alpha1 C). Mechanistically, prenatal DEX exposure resulted in a transcriptionally active chromatin structure at the Cav1.2 gene promoter by altering histone modifications. This activation led to increased expression of vascular Cav1.2 gene, ultimately resulting in increased MCA contractility in offspring. The present study is the first to demonstrate that the adverse effects of prenatal GCs administration on cerebrovascular tone persist into adulthood, providing new insights into developmental origins of cerebrovascular disease. [Display omitted] • Prenatal GCs exposure increased offspring MCA sensitivity to Angiotensin II. • Cav1.2 expression was upregulated in prenatal GCs-exposed offspring MCA. • The increased MCA sensitivity was correlated with upregulated Cav1.2. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Regulation of Cardiac L-type Calcium Channels by 14-3-3 and BIN1
- Author
-
Spooner, Heather
- Subjects
Physiology ,Cellular biology ,14-3-3 ,BIN1 ,CaV1.2 - Abstract
The L-type Ca2+ channel (CaV1.2) is essential for cardiac excitation-contraction coupling. Alterations in CaV1.2 expression, localization, or function can underlie cardiac dysfunction found in aging and heart disease; thus the regulatory mechanisms that tune CaV1.2 expression to meet contractile demand are a crucial area of research. This dissertation work highlights the function of two proteins that regulate CaV1.2 trafficking: 14-3-3 and BIN1. We used a range of techniques including confocal imaging, proximity ligation assays, super-resolution imaging, In vivo echocardiography, and co-immunoprecipitation to explore the dynamics of this regulation. Our work elucidates a novel mechanism where 14-3-3 interacts with CaV1.2 in a phosphorylation-dependent manner to promote enhanced trafficking/recycling, clustering, and activity during β-adrenergic stimulation. We also report that a two-week treatment to restore youthful Bridging Integrator 1 (BIN1) levels in the hearts of 24-month-old mice rejuvenates cardiac function and substantially reverses the dysregulated endosomal recycling and disrupted CaV1.2 trafficking phenotype found in aging. These results increase our understanding of CaV1.2 regulation and provide a foundation for future work establishing a complete model of CaV1.2 trafficking.
- Published
- 2024
21. L- and T-type Ca2+ channels dichotomously contribute to retinal ganglion cell injury in experimental glaucoma
- Author
-
Hong-Ning Wang, Wen-Jing Qian, Guo-Li Zhao, Fang Li, Yan-Ying Miao, Bo Lei, Xing-Huai Sun, and Zhong-Feng Wang
- Subjects
apoptosis ,cav1.2 ,cav3.3 ,chronic ocular hypertension ,extracellular signal-regulated kinase ,mitogen-activated protein kinase ,nuclear factor-kappa b ,patch-clamp ,retina ,tumor necrosis factor-α ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Retinal ganglion cell apoptotic death is the main pathological characteristic of glaucoma, which is the leading cause of irreversible blindness. Disruption of Ca2+ homeostasis plays an important role in glaucoma. Voltage-gated Ca2+ channel blockers have been shown to improve vision in patients with glaucoma. However, whether and how voltage-gated Ca2+ channels are involved in retinal ganglion cell apoptotic death are largely unknown. In this study, we found that total Ca2+ current densities in retinal ganglion cells were reduced in a rat model of chronic ocular hypertension experimental glaucoma, as determined by whole-cell patch-clamp electrophysiological recordings. Further analysis showed that L-type Ca2+ currents were downregulated while T-type Ca2+ currents were upregulated at the later stage of glaucoma. Western blot assay and immunofluorescence experiments confirmed that expression of the CaV1.2 subunit of L-type Ca2+ channels was reduced and expression of the CaV3.3 subunit of T-type Ca2+ channels was increased in retinas of the chronic ocular hypertension model. Soluble tumor necrosis factor-α, an important inflammatory factor, inhibited the L-type Ca2+ current of isolated retinal ganglion cells from control rats and enhanced the T-type Ca2+ current. These changes were blocked by the tumor necrosis factor-α inhibitor XPro1595, indicating that both types of Ca2+ currents may be mediated by soluble tumor necrosis factor-α. The intracellular mitogen-activated protein kinase/extracellular signal-regulated kinase pathway and nuclear factor kappa-B signaling pathway mediate the effects of tumor necrosis factor-α. TUNEL assays revealed that mibefradil, a T-type calcium channel blocker, reduced the number of apoptotic retinal ganglion cells in the rat model of chronic ocular hypertension. These results suggest that T-type Ca2+ channels are involved in disrupted Ca2+ homeostasis and apoptosis of retinal ganglion cells in glaucoma, and application of T-type Ca2+ channel blockers, especially a specific CaV3.3 blocker, may be a potential strategy for the treatment of glaucoma.
- Published
- 2023
- Full Text
- View/download PDF
22. Increased Ca2+ signaling through CaV1.2 induces tendon hypertrophy with increased collagen fibrillogenesis and biomechanical properties.
- Author
-
Li, Haiyin, Korcari, Antonion, Ciufo, David, Mendias, Christopher L., Rodeo, Scott A., Buckley, Mark R., Loiselle, Alayna E., Pitt, Geoffrey S., and Cao, Chike
- Abstract
Tendons are tension‐bearing tissues transmitting force from muscle to bone for body movement. This mechanical loading is essential for tendon development, homeostasis, and healing after injury. While Ca2+ signaling has been studied extensively for its roles in mechanotransduction, regulating muscle, bone, and cartilage development and homeostasis, knowledge about Ca2+ signaling and the source of Ca2+ signals in tendon fibroblast biology are largely unknown. Here, we investigated the function of Ca2+ signaling through CaV1.2 voltage‐gated Ca2+ channel in tendon formation. Using a reporter mouse, we found that CaV1.2 is highly expressed in tendon during development and downregulated in adult homeostasis. To assess its function, we generated ScxCre;CaV1.2TS mice that express a gain‐of‐function mutant CaV1.2 in tendon. We found that mutant tendons were hypertrophic, with more tendon fibroblasts but decreased cell density. TEM analyses demonstrated increased collagen fibrillogenesis in the hypertrophic tendons. Biomechanical testing revealed that the hypertrophic tendons display higher peak load and stiffness, with no changes in peak stress and elastic modulus. Proteomic analysis showed no significant difference in the abundance of type I and III collagens, but mutant tendons had about two‐fold increase in other ECM proteins such as tenascin C, tenomodulin, periostin, type XIV and type VIII collagens, around 11‐fold increase in the growth factor myostatin, and significant elevation of matrix remodeling proteins including Mmp14, Mmp2, and cathepsin K. Taken together, these data highlight roles for increased Ca2+ signaling through CaV1.2 on regulating expression of myostatin growth factor and ECM proteins for tendon collagen fibrillogenesis during tendon formation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. L- and T-type Ca2+ channels dichotomously contribute to retinal ganglion cell injury in experimental glaucoma.
- Author
-
Hong-Ning Wang, Wen-Jing Qian, Guo-Li Zhao, Fang Li, Yan-Ying Miao, Bo Lei, Xing-Huai Sun, and Zhong-Feng Wang
- Published
- 2023
- Full Text
- View/download PDF
24. Wildtype peers rescue social play and 50-kHz ultrasonic vocalization deficits in juvenile female Cacna1c heterozygous rats
- Author
-
Rebecca Bogdan, Rukhshona Kayumova, Rainer K. W. Schwarting, Markus Wöhr, and Theresa M. Kisko
- Subjects
Cav1.2 ,calcium ,rough-and-tumble play ,ultrasonic communication ,social environment ,development ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
BackgroundHealthy brain development depends on early social practices and experiences. The risk gene CACNA1C is implicated in numerous neuropsychiatric disorders, in which key characteristics include deficits in social functioning and communication. Recently, we reported sex-dependent impairments in social behavior and ultrasonic vocalizations (USV) in juvenile heterozygous Cacna1c+/− (HET) rats. Specifically, HET females displayed increases in rough-and-tumble play that eliminated the typically observed sex difference between male and female rats. Interestingly, female wild-type Cacna1c+/+ (WT) pairs also showed a similar increase in social play when housed with HET females, suggesting their behavior may be influenced by HET cage mates. This indicates that the genetic makeup of the social environment related to Cacna1c can influence social play, yet systematic studies are lacking.MethodsIn the present study, we housed juvenile females in MIXED- or SAME-genotype cages and tested them in a social play paradigm with a same- and opposite-genotype partner.ResultsThe results show that the early social environment and the genotype of the play partner influence social play and 50-kHz USV emission. Experience with a WT play partner appears necessary for HET females to show comparable levels of play and 50-kHz USV emission. Same-genotype HET pairs played less and emitted fewer 50-kHz USV than same-genotype WT or opposite-genotype pairs; however, we found that the decrease in social play and 50-kHz USV in HET pairs can be rescued by playing with a WT partner. The effect was particularly prominent when the first play partner was WT, as we found it increased play and 50-kHz USV emission in all subsequent interactions with ensuing partners.ConclusionThese findings suggest that the genetic makeup related to the social environment and/or social peers influences social play in Cacna1c+/− haploinsufficient rats. Specifically, our results show that WT peers can rescue behavior and communication alterations in Cacna1c female rats. Our findings have important implications because they show that the genetic makeup of the social environment can divulge phenotypic changes in genetic rat models of neuropsychiatric disorders.
- Published
- 2023
- Full Text
- View/download PDF
25. Functional Role and Plasticity of Voltage-Gated Calcium Channels in the Control of Heart Automaticity
- Author
-
Mesirca, Pietro, Bidaud, Isabelle, Torre, Eleonora, Torrente, Angelo G., D’Souza, Alicia, Mangoni, Matteo E., Zamponi, Gerald Werner, editor, and Weiss, Norbert, editor
- Published
- 2022
- Full Text
- View/download PDF
26. Chemical shift assignments of a calmodulin intermediate with two Ca2+ bound in complex with the IQ-motif of voltage-gated Ca2+ channels (CaV1.2)
- Author
-
Salveson, Ian, Anderson, David E, Hell, Johannes W, and Ames, James B
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Amides ,Amino Acid Motifs ,Calcium ,Calcium Channels ,Calmodulin ,Humans ,Nuclear Magnetic Resonance ,Biomolecular ,Protein Binding ,Protein Structure ,Secondary ,EF-hand ,Ca(V)1 ,2 ,IQ-motif ,Synaptic plasticity ,CDI ,CaV1.2 ,Biophysics ,Biochemistry and cell biology - Abstract
Calcium-dependent inactivation (CDI) of neuronal voltage-gated Ca2+ channels (CaV1.2) is important for synaptic plasticity, which is associated with learning and memory. The Ca2+-dependent binding of calmodulin (CaM) to CaV1.2 is essential for CDI. Here we report NMR assignments for a CaM mutant (D21A/D23A/D25A/E32Q/D57A/D59A/N61A/E68Q, called CaMEF12) that contains two Ca2+ bound at the third and fourth EF-hands (EF3 and EF4) and is bound to the IQ-motif (residues 1644-1665) from CaV1.2 (BMRB accession no. 27692).
- Published
- 2019
27. Unraveled roles of Cav1.2 in proliferation and stemness of ameloblastoma
- Author
-
Shujin Li, Dong-Joon Lee, Hyun-Yi Kim, Jun-Young Kim, Young-Soo Jung, and Han-Sung Jung
- Subjects
Ameloblastoma ,Proliferation ,Stemness ,Calcium signaling ,Cav1.2 ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Transcriptome analysis has been known as a functional tool for cancer research recently. Mounting evidence indicated that calcium signaling plays several key roles in cancer progression. Despite numerous studies examining calcium signaling in cancer, calcium signaling studies in ameloblastoma are limited. Results In the present study, comparative transcriptome profiling of two representative odontogenic lesions, ameloblastoma and odontogenic keratocyst, revealed that Cav1.2 (CACNA1C, an L-type voltage-gated calcium channel) is strongly enriched in ameloblastoma. It was confirmed that the Ca2+ influx in ameloblastoma cells is mainly mediated by Cav1.2 through L-type voltage-gated calcium channel agonist and blocking reagent treatment. Overexpression and knockdown of Cav1.2 showed that Cav1.2 is directly involved in the regulation of the nuclear translocation of nuclear factor of activated T cell 1 (NFATc1), which causes cell proliferation. Furthermore, a tumoroid study indicated that Cav1.2-dependent Ca2+ entry is also associated with the maintenance of stemness of ameloblastoma cells via the enhancement of Wnt/β-catenin signaling activity. Conclusion In conclusion, Cav1.2 regulates the NFATc1 nuclear translocation to enhance ameloblastoma cell proliferation. Furthermore, Cav1.2 dependent Ca2+ influx contributes to the Wnt/β-catenin activity for the ameloblastoma cell stemness and tumorigenicity. Our fundamental findings could have a major impact in the fields of oral maxillofacial surgery, and genetic manipulation or pharmacological approaches to Cav1.2 can be considered as new therapeutic options.
- Published
- 2022
- Full Text
- View/download PDF
28. Regulation of Cardiac Cav1.2 Channels by Calmodulin.
- Author
-
Kameyama, Masaki, Minobe, Etsuko, Shao, Dongxue, Xu, Jianjun, Gao, Qinghua, and Hao, Liying
- Subjects
- *
CALMODULIN , *PHOSPHOPROTEIN phosphatases , *PROTEIN kinases , *CALCINEURIN , *ENZYMES - Abstract
Cav1.2 Ca2+ channels, a type of voltage-gated L-type Ca2+ channel, are ubiquitously expressed, and the predominant Ca2+ channel type, in working cardiac myocytes. Cav1.2 channels are regulated by the direct interactions with calmodulin (CaM), a Ca2+-binding protein that causes Ca2+-dependent facilitation (CDF) and inactivation (CDI). Ca2+-free CaM (apoCaM) also contributes to the regulation of Cav1.2 channels. Furthermore, CaM indirectly affects channel activity by activating CaM-dependent enzymes, such as CaM-dependent protein kinase II and calcineurin (a CaM-dependent protein phosphatase). In this article, we review the recent progress in identifying the role of apoCaM in the channel 'rundown' phenomena and related repriming of channels, and CDF, as well as the role of Ca2+/CaM in CDI. In addition, the role of CaM in channel clustering is reviewed. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
29. 'Toxic Masculinity': What Is Known about the Role of Androgen Receptors in Head and Neck Squamous Cell Carcinoma.
- Author
-
Čonkaš, Josipa, Sabol, Maja, and Ozretić, Petar
- Subjects
- *
SQUAMOUS cell carcinoma , *POISONS , *NUCLEAR membranes , *HUMAN papillomavirus , *HEAD & neck cancer , *ANDROGEN receptors , *NECK , *CETUXIMAB - Abstract
Head and neck squamous cell carcinoma (HNSCC), the most prevalent cancer in the head and neck region, develops from the mucosal epithelium of the upper aerodigestive tract. Its development directly correlates with alcohol and/or tobacco consumption and infection with human papillomavirus. Interestingly, the relative risk for HNSCC is up to five times higher in males, so it is considered that the endocrine microenvironment is another risk factor. A gender-specific risk for HNSCC suggests either the existence of specific risk factors that affect only males or that females have defensive hormonal and metabolic features. In this review, we summarized the current knowledge about the role of both nuclear and membrane androgen receptors (nAR and mARs, respectively) in HNSCC. As expected, the significance of nAR is much better known; it was shown that increased nAR expression was observed in HNSCC, while treatment with dihydrotestosterone increased proliferation, migration, and invasion of HNSCC cells. For only three out of five currently known mARs—TRPM8, CaV1.2, and OXER1—it was shown either their increased expression in various types of HNSCC or that their increased activity enhanced the migration and invasion of HNSCC cells. The primary treatments for HNSCC are surgery and radiotherapy, but targeted immunotherapies are on the rise. On the other hand, given the evidence of elevated nAR expression in HNSCC, this receptor represents a potential target for antiandrogen therapy. Moreover, there is still plenty of room for further examination of mARs' role in HNSCC diagnosis, prognosis, and treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Suxiao Jiuxin Pills Prevent Ventricular Fibrillation from Inhibiting L-type Calcium Currents CaV1.2 in vivo and in vitro.
- Author
-
Qi, Jian-yong, Kang, Dong-yuan, Yu, Juan, and Zhang, Min-zhou
- Subjects
IN vitro studies ,BIOLOGICAL models ,COMPUTER simulation ,VERAPAMIL ,HERBAL medicine ,IN vivo studies ,CALCIUM antagonists ,ANIMAL experimentation ,CONVALESCENCE ,RABBITS ,POTASSIUM ,CELL physiology ,TREATMENT effectiveness ,ELECTROCARDIOGRAPHY ,VENTRICULAR fibrillation ,CYTOLOGY ,CHINESE medicine ,MICE - Abstract
Objective: To investigate whether Suxiao Jiuxin Pills (SJP), a Chinese herbal remedy, is an anti-ventricular fibrillation (VF) agent. Methods: VF was induced by isoproterenolol (ISO) intraperitoneal injection followed by electrical pacing in mice and rabbits. The effects of SJP on the L-type calcium channel current (CaV1.2), voltage-dependent sodium channel current (I
Na ), rapid and slow delayed rectifier potassium channel current (IKr and IKs , respectively) were studied by whole-cell patch-clamp method. Computer simulation was implemented to incorporate the experimental data of SJP effects on the CaV1.2 current into the action potential (AP) and pseudo-electrocardiography (pseudo-ECG) models. Results: SJP prevented VF induction and reduced VF durations significantly in mice and rabbits. Patch-clamp experiments revealed that SJP decreased the peak amplitude of the CaV1.2 current with a half maximal concentration (IC50 ) value of 16.9 mg/L (SJP-30 mg/L, −32.8 ± 6.1 pA; Verapamil, −16.2 ±1.8 pA; vs. control, −234.5 ±16.7 pA, P<0.01, respectively). The steady-state activation curve, inactivation curve, and the recovery from inactivation of the CaV1.2 current were not shifted significantly. Specifically, SJP did not altered INa , IKr , and IKs currents significantly (SJP vs. control, P>0.05). Computer simulation showed that SJP-reduced CaV1.2 current shortened the AP duration, transiting VF into sinus rhythm in pseudo-ECG. Conclusion: SJP reduced VF via inhibiting the CaV1.2 current with in vivo, in vitro, and in silico studies, which provide experimental basis for SJP anti-VF clinical application. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
31. BIN1 Induces the Formation of T-Tubules and Adult-Like Ca2+ Release Units in Developing Cardiomyocytes
- Author
-
De La Mata, Ana, Tajada, Sendoa, O'Dwyer, Samantha, Matsumoto, Collin, Dixon, Rose E, Hariharan, Nirmala, Moreno, Claudia M, and Santana, Luis Fernando
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Stem Cell Research - Embryonic - Human ,Stem Cell Research ,Regenerative Medicine ,Heart Disease ,Cardiovascular ,5.2 Cellular and gene therapies ,Adaptor Proteins ,Signal Transducing ,Calcium ,Calcium Signaling ,Cell Differentiation ,Humans ,Myocytes ,Cardiac ,Nuclear Proteins ,Tumor Suppressor Proteins ,hESC ,Cardiac myocytes ,BIN1 ,T-tubules ,Ca(V)1.2 ,Calcium release units ,CaV1.2 ,Biological Sciences ,Technology ,Medical and Health Sciences ,Immunology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) are at the center of new cell-based therapies for cardiac disease, but may also serve as a useful in vitro model for cardiac cell development. An intriguing feature of hESC-CMs is that although they express contractile proteins and have sarcomeres, they do not develop transverse-tubules (T-tubules) with adult-like Ca2+ release units (CRUs). We tested the hypothesis that expression of the protein BIN1 in hESC-CMs promotes T-tubules formation, facilitates CaV 1.2 channel clustering along the tubules, and results in the development of stable CRUs. Using electrophysiology, [Ca2+ ]i imaging, and super resolution microscopy, we found that BIN1 expression induced T-tubule development in hESC-CMs, while increasing differentiation toward a more ventricular-like phenotype. Voltage-gated CaV 1.2 channels clustered along the surface sarcolemma and T-tubules of hESC-CM. The length and width of the T-tubules as well as the expression and size of CaV 1.2 clusters grew, as BIN1 expression increased and cells matured. BIN1 expression increased CaV 1.2 channel activity and the probability of coupled gating within channel clusters. Interestingly, BIN1 clusters also served as sites for sarcoplasmic reticulum (SR) anchoring and stabilization. Accordingly, BIN1-expressing cells had more CaV 1.2-ryanodine receptor junctions than control cells. This was associated with larger [Ca2+ ]i transients during excitation-contraction coupling. Our data support the view that BIN1 is a key regulator of T-tubule formation and CaV 1.2 channel delivery. By studying the role of BIN1 during the differentiation of hESC-CMs, we show that BIN1 is also important for CaV 1.2 channel clustering, junctional SR organization, and the establishment of excitation-contraction coupling. Stem Cells 2019;37:54-64.
- Published
- 2019
32. Proteolytic processing of the L-type Ca 2+ channel alpha 11.2 subunit in neurons
- Author
-
Buonarati, Olivia R, Henderson, Peter B, Murphy, Geoffrey G, Horne, Mary C, and Hell, Johannes W
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Neurosciences ,Biotechnology ,Genetics ,1.1 Normal biological development and functioning ,Neurological ,Cav1.2 ,calpain cleavage ,neuronal calcium ,Clinical Sciences ,Oncology and Carcinogenesis - Abstract
Background: The L-type Ca2+ channel Cav1.2 is a prominent regulator of neuronal excitability, synaptic plasticity, and gene expression. The central element of Cav1.2 is the pore-forming α 11.2 subunit. It exists in two major size forms, whose molecular masses have proven difficult to precisely determine. Recent work suggests that α 11.2 is proteolytically cleaved between the second and third of its four pore-forming domains (Michailidis et al,. 2014). Methods: To better determine the apparent molecular masses (M R)of the α 11.2 size forms, extensive systematic immunoblotting of brain tissue as well as full length and C-terminally truncated α 11.2 expressed in HEK293 cells was conducted using six different region-specific antibodies against α 11.2. Results: The full length form of α 11.2 migrated, as expected, with an apparent M R of ~250 kDa. A shorter form of comparable prevalence with an apparent M R of ~210 kDa could only be detected in immunoblots probed with antibodies recognizing α 11.2 at an epitope 400 or more residues upstream of the C-terminus. Conclusions: The main two size forms of α 11.2 are the full length form and a shorter form, which lacks ~350 distal C-terminal residues. Midchannel cleavage as suggested by Michailidis et al. (2014) is at best minimal in brain tissue.
- Published
- 2018
33. The rs216009 single-nucleotide polymorphism of the CACNA1C gene is associated with phantom tooth pain.
- Author
-
Masako Morii, Seii Ohka, Daisuke Nishizawa, Junko Hasegawa, Kyoko Nakayama, Yuko Ebata, Moe Soeda, Ken-ichi Fukuda, Kaori Yoshida, Kyotaro Koshika, Tatsuya Ichinohe, and Kazutaka Ikeda
- Subjects
- *
SINGLE nucleotide polymorphisms , *GENETIC polymorphisms , *RECESSIVE genes , *RECOLLECTION (Psychology) , *NEURALGIA , *AFFECTIVE neuroscience , *TOOTHACHE - Abstract
Phantom tooth pain (PTP) is a rare and specific neuropathic pain that occurs after pulpectomy and tooth extraction, but its cause is not understood. We hypothesized that there is a genetic contribution to PTP. The present study focused on the CACNA1C gene, which encodes the α1C subunit of the Cav1.2 L-type Ca2+ channel (LTCC) that has been reported to be associated with neuropathic pain in previous studies. We investigated genetic polymorphisms that contribute to PTP. We statistically examined the association between genetic polymorphisms and PTP vulnerability in 33 patients with PTP and 118 patients without PTP but with pain or dysesthesia in the orofacial region. From within and around the CACNA1C gene, 155 polymorphisms were selected and analyzed for associations with clinical data. We found that the rs216009 single-nucleotide polymorphism (SNP) of the CACNA1C gene in the recessive model was significantly associated with the vulnerability to PTP. Homozygote carriers of the minor C allele of rs216009 had a higher rate of PTP. Nociceptive transmission in neuropathic pain has been reported to involve Ca2+ influx from LTCCs, and the rs216009 polymorphism may be involved in CACNA1C expression, which regulates intracellular Ca2+ levels, leading to the vulnerability to PTP. Furthermore, psychological factors may lead to the development of PTP by modulating the descending pain inhibitory system. Altogether, homozygous C-allele carriers of the rs216009 SNP were more likely to be vulnerable to PTP, possibly through the regulation of intracellular Ca2+ levels and affective pain systems, such as those that mediate fear memory recall. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
34. Cav1.2 regulated odontogenic differentiation of NG2+ pericytes during pulp injury.
- Author
-
Fu, Yunyu, Ju, Yanqin, and Zhao, Shouliang
- Subjects
PERICYTES ,MESENCHYMAL stem cells ,DENTAL pulp ,POLYMERASE chain reaction ,TRANSGENIC mice - Abstract
NG2
+ pericytes, as the possible precursor cells of mesenchymal stem cells (MSCs), have drawn attention due to their ability to differentiate into odontoblasts. Cav 1.2 is involved in the differentiation process of stem cells, but its role in the differentiation of NG2+ pericytes is not clear. The aim of the present study was to examine the role of Cav 1.2 in the differentiation of NG2+ pericytes into odontoblasts. NG2+ pericytes were obtained from human dental pulp cells by magnetic-activated cell sorting. During the odontogenic differentiation of NG2+ pericytes, the effects of the Cav 1.2 inhibitors, nimodipine and Cav 1.2 knockdown shRNA, were analyzed by real-time polymerase chain reaction and alizarin red staining. NG2CreERT2/Rosa26-GFP lineage-tracing mice were established to further investigate the roles of NG2+ pericytes and Cav 1.2 in incisor self-repair after injury in vivo. At 10 min, 1 day, and 3 days after pulp injuries in transgenic mice, NG2-GFP+ and Cav 1.2 immunofluorescence co-staining was performed on the incisors. Nimodipine treatment and Cav 1.2 knockdown showed similar inhibition of calcium nodule formation and mRNA levels of osteogenic markers (DSPP, DMP1, and Runx2, p < 0.05). NG2+ pericytes migrated from their inherent perivascular location to the odontoblast layers after pulp injury. Cav 1.2 showed a similar response pattern as NG2+ pericytes and gradually returned to normal levels. In addition, many co-stained areas of Cav 1.2 and NG2+ pericytes, both near the perivascular and odontoblast layers, were observed. These results indicate that Cav 1.2 played a vital role in the odontogenic differentiation of NG2+ pericytes, and that it might be closely linked to the NG2+ pericytes-mediated repair of dental pulp injury in vivo. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
35. Oxidative Regulation of Vascular Ca v 1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders.
- Author
-
Hu, Xiang-Qun and Zhang, Lubo
- Subjects
VASCULAR resistance ,BLOOD pressure ,PREECLAMPSIA ,HYPERTENSION ,CALCIUM channels ,REACTIVE oxygen species ,CARDIAC output - Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca
2+ (Cav 1.2) channel in small arteries and arterioles plays an essential role in regulating Ca2+ influx, vascular resistance, and blood pressure. Hypertension and preeclampsia are characterized by high blood pressure. In addition, diabetes has a high prevalence of hypertension. The etiology of these disorders remains elusive, involving the complex interplay of environmental and genetic factors. Common to these disorders are oxidative stress and vascular dysfunction. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria are primary sources of vascular oxidative stress, whereas dysfunction of the Cav 1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav 1.2 and potential roles of ROS-mediated Cav 1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
36. Calmodulin promotes a Ca2+‐dependent conformational change in the C‐terminal regulatory domain of CaV1.2.
- Author
-
Yadav, Deepak Kumar, Anderson, David E., Hell, Johannes W., and Ames, James B.
- Subjects
- *
CALMODULIN , *CALCIUM ions - Abstract
Calmodulin (CaM) binds to the membrane‐proximal cytosolic C‐terminal domain of CaV1.2 (residues 1520–1669, CT(1520–1669)) and causes Ca2+‐induced conformational changes that promote Ca2+‐dependent channel inactivation (CDI). We report biophysical studies that probe the structural interaction between CT(1520–1669) and CaM. The recombinantly expressed CT(1520–1669) is insoluble, but can be solubilized in the presence of Ca2+‐saturated CaM (Ca4/CaM), but not in the presence of Ca2+‐free CaM (apoCaM). We show that half‐calcified CaM (Ca2/CaM12) forms a complex with CT(1520–1669) that is less soluble than CT(1520–1669) bound to Ca4/CaM. The NMR spectrum of CT(1520–1669) reveals spectral differences caused by the binding of Ca2/CaM12 versus Ca4/CaM, suggesting that the binding of Ca2+ to the CaM N‐lobe may induce a conformational change in CT(1520–1669). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
37. Calcium Channel Splice Variants and Their Effects in Brain and Cardiovascular Function
- Author
-
Yeow, Sean Qing Zhang, Loh, Kelvin Wei Zhern, Soong, Tuck Wah, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Steinlein, Ortrud, Series Editor, Xiao, Junjie, Series Editor, and Zhou, Lei, editor
- Published
- 2021
- Full Text
- View/download PDF
38. Evaluation of four KCNMA1 channelopathy variants on BK channel current under Ca V 1.2 activation.
- Author
-
Dinsdale RL and Meredith AL
- Subjects
- Humans, HEK293 Cells, Channelopathies genetics, Channelopathies metabolism, Calcium metabolism, Large-Conductance Calcium-Activated Potassium Channels metabolism, Large-Conductance Calcium-Activated Potassium Channels genetics, Mutation, Large-Conductance Calcium-Activated Potassium Channel alpha Subunits genetics, Large-Conductance Calcium-Activated Potassium Channel alpha Subunits metabolism, Calcium Channels, L-Type metabolism, Calcium Channels, L-Type genetics
- Abstract
Variants in KCNMA1 , encoding the voltage- and calcium-activated K
+ (BK) channel, are associated with human neurological disease. The effects of gain-of-function (GOF) and loss-of-function (LOF) variants have been predominantly studied on BK channel currents evoked under steady-state voltage and Ca2+ conditions. However, in their physiological context, BK channels exist in partnership with voltage-gated Ca2+ channels and respond to dynamic changes in intracellular Ca2+ (Ca2+ i ). In this study, an L-type voltage-gated Ca2+ channel present in the brain, CaV 1.2, was co-expressed with wild type and mutant BK channels containing GOF (D434G, N999S) and LOF (H444Q, D965V) patient-associated variants in HEK-293T cells. Whole-cell BK currents were recorded under CaV 1.2 activation using buffering conditions that restrict Ca2+ i to nano- or micro-domains. Both conditions permitted wild type BK current activation in response to CaV 1.2 Ca2+ influx, but differences in behavior between wild type and mutant BK channels were reduced compared to prior studies in clamped Ca2+ i . Only the N999S mutation produced an increase in BK current in both micro- and nano-domains using square voltage commands and was also detectable in BK current evoked by a neuronal action potential within a microdomain. These data corroborate the GOF effect of N999S on BK channel activity under dynamic voltage and Ca2+ stimuli, consistent with its pathogenicity in neurological disease. However, the patient-associated mutations D434G, H444Q, and D965V did not exhibit significant effects on BK current under CaV 1.2-mediated Ca2+ influx, in contrast with prior steady-state protocols. These results demonstrate a differential potential for KCNMA1 variant pathogenicity compared under diverse voltage and Ca2+ conditions.- Published
- 2024
- Full Text
- View/download PDF
39. A vascular smooth muscle-specific integrin-α8 Cre mouse for lymphatic contraction studies that allows male-female comparisons and avoids visceral myopathy
- Author
-
Michael J. Davis, Hae Jin Kim, Min Li, and Scott D. Zawieja
- Subjects
lymphatic smooth muscle ,Myh11-CreERT2 ,Itga8-CreERT2 ,integrin alpha-8 ,L-type voltage-gated calcium channel ,CaV1.2 ,Physiology ,QP1-981 - Abstract
Introduction: The widely-used, tamoxifen-inducible, smooth muscle (SM)-specific Cre, Myh11-CreERT2, suffers from two disadvantages: 1) it is carried on the Y-chromosome and thus only effective for gene deletion in male mice, and 2) it recombines in both vascular and non-vascular SM, potentially leading to unwanted or confounding gastrointestinal phenotypes. Here, we tested the effectiveness of a new, SM-specific Cre, based on the integrin α8 promoter (Itga8-CreERT2), that has been recently developed and characterized, to assess the effects of Cav1.2 deletion on mouse lymphatic SM function.Methods:Cav1.2 (the L-type voltage-gated calcium channel) is essential for lymphatic pacemaking and contraction and its deletion using either Myh11-CreERT2 or Itga8-CreERT2 abolished spontaneous lymphatic contractions. Mouse lymphatic contractile function was assessed using two ex vivo methods.Results:Myh11-CreERT2; Cav1.2f/f mice died of gastrointestinal obstruction within 20 days of the first tamoxifen injection, preceded by several days of progressively poor health, with symptoms including weight loss, poor grooming, hunched posture, and reduced overall activity. In contrast, Itga8-CreERT2; Cav1.2f/f mice survived for >80 days after induction and were in normal health until the time of sacrifice for experimental studies. Cav1.2 deletion was equally effective in male and female mice.Discussion: Our results demonstrate that Itga8-CreERT2 can be used to effectively delete genes in lymphatic smooth muscle while avoiding potentially lethal visceral myopathy and allowing comparative studies of lymphatic contractile function in both male and female mice.
- Published
- 2023
- Full Text
- View/download PDF
40. Ion Channel Clusters in Arterial Smooth Muscle: Investigating the Mechanisms of KV2.1 and CaV1.2 Formation in Murine Mesenteric Cells and Their Implications for Smooth Muscle Physiology and Sex Differences
- Author
-
Matsumoto, Collin Scott Tadashi
- Subjects
Physiology ,Arterial smooth muscle ,CaV1.2 ,Ion channel clustering ,KV2.1 ,Sex-differences - Abstract
The work contained within this dissertation aims to investigate the role and mechanisms of CaV1.2 and KV2.1 ion channel cluster formation in the membrane of arterial smooth muscle cells. Clustering of ion channels selective for these ions have long been a commonly observed phenotype, yet the mechanisms of cluster formation are still unknown. Overall, this dissertation provides a model by which ion channel clusters form stochastically via a self-assembly process in the membrane based on three mechanistic probabilities: nucleation, growth, and removal. Additionally, we focused on clustering and interactions of two key ion channels, KV2.1 and CaV1.2, in smooth muscle physiology selective for potassium (K+) and calcium (Ca2+) respectively. The opening of these channels play key roles in arterial physiology, counterbalancing each other to affect arterial diameter. The key findings from this work help to elucidate the mechanisms involved in the trafficking of ion channels, maintenance of clusters in the plasma membrane and provide potential reasoning for sex-based differences in smooth muscle physiology. Using a multiscale experimental and computational approach, we describe a key interaction between CaV1.2 and KV2.1 in arterial smooth muscle. This model proposes that KV2.1 clustering state is not a determinate of channel conduction in mesenteric smooth muscle. Additionally, KV2.1 macro-clusters serve as a sex-specific site for increased CaV1.2 clustering and decreasing KV2.1 macro-clustering decreases CaV1.2 channel clustering. This sex-based interaction ultimately plays a key role in Ca2+ dynamics and smooth muscle physiology.
- Published
- 2023
41. Convergent regulation of CaV1.2 channels by direct phosphorylation and by the small GTPase RAD in the cardiac fight-or-flight response.
- Author
-
Hovey, Liam, Gamal El-Din, Tamer M., and Catterall, William A.
- Subjects
- *
GUANOSINE triphosphatase , *PHOSPHORYLATION , *CALCIUM channels , *HEART beat - Abstract
The L-type calcium currents conducted by the cardiac CaV1.2 calcium channel initiate excitation–contraction coupling and serve as a key regulator of heart rate, rhythm, and force of contraction. CaV1.2 is regulated by β-adrenergic/protein kinase A (PKA)- mediated protein phosphorylation, proteolytic processing, and autoinhibition by its carboxyl-terminal domain (CT). The small guanosine triphosphatase (GTPase) RAD (Ras associated with diabetes) has emerged as a potent inhibitor of CaV1.2, and accumulating evidence suggests a key role for RAD in mediating β-adrenergic/PKA upregulation of channel activity. However, the relative roles of direct phosphorylation of CaV1.2 channels and phosphorylation of RAD in channel regulation remain uncertain. Here, we investigated the hypothesis that these two mechanisms converge to regulate CaV1.2 channels. Both RAD and the proteolytically processed distal CT (dCT) strongly reduced CaV1.2 activity. PKA phosphorylation of RAD and phosphorylation of Ser- 1700 in the proximal CT (pCT) synergistically reversed this inhibition and increased CaV1.2 currents. Our findings reveal that the proteolytically processed form of CaV1.2 undergoes convergent regulation by direct phosphorylation of the CT and by phosphorylation of RAD. These parallel regulatory pathways provide a flexible mechanism for upregulation of the activity of CaV1.2 channels in the fight-or-flight response. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
42. Sites and Regulation of L-Type Ca2+ Channel Cav1.2 Phosphorylation in Brain.
- Author
-
Shin, Seok Kyo, Li, Hai Ying, Cho, Kun, Cho, Young Wuk, Lee, Jung-Ha, and Park, Kang-Sik
- Subjects
- *
CALCIUM channels , *PHOSPHORYLATION , *MYOCARDIUM , *NEUROPLASTICITY , *MASS spectrometry - Abstract
Cav1.2 channel phosphorylation plays an important role in regulating neuronal plasticity by action potential-dependent Ca2+ entry. Most studies of Cav1.2 regulation by phosphorylation have been reported in heart and muscles. Here, we identified phosphorylation sites of neuronal Cav1.2 channel protein purified from rat brain using mass spectrometry. The functional characterization of these phosphorylation sites showed altered voltage-dependent biophysical properties of the channel, without affecting current density. These results show that neuronal Cav1.2 channel is regulated by phosphorylation in a complex mechanism involving multiple phosphorylation sites. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
43. Chemical shift assignments of the C-terminal domain of CaBP1 bound to the IQ-motif of voltage-gated Ca2+ channel (CaV1.2).
- Author
-
Salveson, Ian and Ames, James B.
- Abstract
The neuronal L-type voltage-gated Ca
2+ channel (CaV 1.2) interacts with Ca2+ binding protein 1 (CaBP1), that promotes Ca2+ -induced channel activity. The binding of CaBP1 to the IQ-motif in CaV 1.2 (residues 1644–1665) blocks the binding of calmodulin and prevents Ca2+ -dependent inactivation of CaV 1.2. This Ca2+ -induced binding of CaBP1 to CaV 1.2 is important for modulating neuronal synaptic plasticity, which may serve a role in learning and memory. Here we report NMR assignments of the C-terminal domain of CaBP1 (residues 99–167, called CaBP1C) that contains two Ca2+ bound at the third and fourth EF-hands (EF3 and EF4) and is bound to the CaV 1.2 IQ-motif from CaV 1.2 (BMRB accession no. 51518). [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
44. Unraveled roles of Cav1.2 in proliferation and stemness of ameloblastoma.
- Author
-
Li, Shujin, Lee, Dong-Joon, Kim, Hyun-Yi, Kim, Jun-Young, Jung, Young-Soo, and Jung, Han-Sung
- Subjects
- *
CALCIUM channels , *AMELOBLASTOMA , *MAXILLOFACIAL surgery , *T cells , *ORAL surgery , *CANCER invasiveness - Abstract
Background: Transcriptome analysis has been known as a functional tool for cancer research recently. Mounting evidence indicated that calcium signaling plays several key roles in cancer progression. Despite numerous studies examining calcium signaling in cancer, calcium signaling studies in ameloblastoma are limited. Results: In the present study, comparative transcriptome profiling of two representative odontogenic lesions, ameloblastoma and odontogenic keratocyst, revealed that Cav1.2 (CACNA1C, an L-type voltage-gated calcium channel) is strongly enriched in ameloblastoma. It was confirmed that the Ca2+ influx in ameloblastoma cells is mainly mediated by Cav1.2 through L-type voltage-gated calcium channel agonist and blocking reagent treatment. Overexpression and knockdown of Cav1.2 showed that Cav1.2 is directly involved in the regulation of the nuclear translocation of nuclear factor of activated T cell 1 (NFATc1), which causes cell proliferation. Furthermore, a tumoroid study indicated that Cav1.2-dependent Ca2+ entry is also associated with the maintenance of stemness of ameloblastoma cells via the enhancement of Wnt/β-catenin signaling activity. Conclusion: In conclusion, Cav1.2 regulates the NFATc1 nuclear translocation to enhance ameloblastoma cell proliferation. Furthermore, Cav1.2 dependent Ca2+ influx contributes to the Wnt/β-catenin activity for the ameloblastoma cell stemness and tumorigenicity. Our fundamental findings could have a major impact in the fields of oral maxillofacial surgery, and genetic manipulation or pharmacological approaches to Cav1.2 can be considered as new therapeutic options. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
45. Nuciferine analogs block voltage-gated sodium, calcium and potassium channels to regulate the action potential and treat arrhythmia.
- Author
-
Zhou, Ying Xun, Wang, Wen Ping, Ke, Jin, Ou, Hui Ping, Chen, Lin Yun, Hou, An Guo, Li, Peng, Ma, Yun Shu, and Bin Jin, Wen
- Subjects
- *
PROARRHYTHMIA , *ION channels , *POTASSIUM channels , *HEART failure , *CALCIUM channels - Abstract
Dysfunction of the Nav1.5, Cav1.2, and Kv channels could interfere with the AP and result in arrhythmias and even heart failure. We herein present a novel library of nuciferine analogs that target ion channels for the treatment of arrhythmias. Patch clamp measurements of ventricular myocytes revealed that 6a dramatically blocked both the I Na and I Ca without altering the current voltage relationship (including the activation potential and peak potential), accelerated the inactivation of Nav and Cav channels and delayed the resurrection of these channels after inactivation. Additionally, 6a significantly decreased the APA and RMP without affecting the APD30 or APD50. The IC 50 values of 6a against Nav1.5 and Cav1.2 were 4.98 μM and 4.62 μM, respectively. Furthermore, 6a (10 μM) blocked I Ks , I K1 , and I to with values of 17.01 %±2.54 %, 9.09 %±2.78 %, and 11.15 %±3.52 %, respectively. Surprisingly, 6a weakly inhibited hERG channels, suggesting a low risk of proarrhythmia. The cytotoxicity evaluation of 6a with the H9c2 cell line indicated that this compound was noncytotoxic. In vivo studies suggested that these novel nuciferine analogs could shorten the time of arrhythmia continuum induced by BaCl 2 and normalize the HR, QRS, QT and QTc interval and the R wave amplitude. Moreover, 6a dose-dependently affected aconitine-induced arrhythmias and notably improved the cumulative dosage of aconitine required to evoke VP, VT, VF and CA in rats with aconitine-induced arrhythmia. In conclusion, nuciferine analogs could be promising ion channel blockers that could be further developed into antiarrhythmic agents. [Display omitted] • A novel library of nuciferine analogs that target ion channels for the treatment of arrhythmias have been developed. • 6a dramatically blocked both the I Na and I Ca without altering the current‒voltage relationship. • 6a weakly inhibited hERG channels, suggesting a low risk of proarrhythmia. • Nuciferine analogs could shorten the time of arrhythmia continuum induced by BaCl 2 and normalize the ECG parameters. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Cellular mechanism underlying the facilitation of contractile response induced by IL-25 in mouse tracheal smooth muscle.
- Author
-
Ze-Xin Huang, Zhuo-Er Qiu, Lei Chen, Xiao-Chun Hou, Yun-Xin Zhu, Wen-Liang Zhou, and Yi-Lin Zhang
- Subjects
- *
SMOOTH muscle , *SMOOTH muscle contraction , *MICE , *MUSCLE cells , *EPITHELIAL cells , *BRONCHIAL spasm - Abstract
Asthma is a common heterogeneous respiratory disease characterized by airway inflammation and airway hyperresponsiveness (AHR) which is associated with abnormality in smooth muscle contractility. The epithelial cell-derived cytokine IL-25 is implicated in type 2 immune pathology including asthma, whereas the underlying mechanisms have not been fully elucidated. This study aims to investigate the effects of IL-25 on mouse tracheal smooth muscle contractility and elucidate the cellular mechanisms. Incubation with IL-25 augmented the contraction of mouse tracheal smooth muscles, which could be suppressed by the L-type voltage-dependent Ca2+ channel (L-VDCC) blocker nifedipine. Furthermore, IL-25 enhanced the cytosolic Ca2+ signals and triggered the upregulation of a1C L-VDCC (CaV1.2) in primary cultured mouse tracheal smooth muscle cells. Knocking down IL-17RA/IL-17RB receptors or inhibiting the transforming growth factor-b-activated kinase 1 (TAK1)-tumor progression locus 2 (TPL2)-MAPK kinase 1/2 (MEK1/2)-ERK1/2-activating protein-1 (AP-1) signaling pathways suppressed the IL-25-elicited upregulation of CaV1.2 and hyperreactivity in tracheal smooth muscles. Moreover, inhibition of TPL2, ERK1/2 or L-VDCC alleviated the AHR symptom induced by IL-25 in a murine model. This study revealed that IL-25 potentiated the contraction of tracheal smooth muscle and evoked AHR via activation of TPL2-ERK1/2-CaV1.2 signaling, providing novel targets for the treatment of asthma with a high-IL-25 phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. Chemical shift assignments of the C-terminal EF-hand domain of α-actinin-1
- Author
-
Turner, Matthew, Anderson, David E, Rajan, Sahana, Hell, Johannes W, and Ames, James B
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Actinin ,Amino Acid Sequence ,EF Hand Motifs ,Humans ,Nuclear Magnetic Resonance ,Biomolecular ,alpha-Actinin-1 ,EF-hand ,Ca(V)1.2 ,IQ-motif ,Synaptic plasticity ,Long-term depression ,CaV1.2 ,α-Actinin-1 ,Biophysics ,Biochemistry and cell biology - Abstract
The regulation and localization of the neuronal voltage gated Ca(2+) channel CaV1.2 is important for synaptic plasticity associated with learning and memory. The cytoskeletal protein, α-actinin-1 is known to interact with CaV1.2 and stabilize its localization at the postsynaptic membrane. Here we report both backbone and sidechain NMR assignments for the C-terminal EF-hands (EF3 and EF4) of α-actinin-1 (residues 824-892, called ACTN_EF34) bound to the IQ-motif (residues 1644-1665) from CaV1.2 (BMRB accession no. 25902).
- Published
- 2016
48. Calcium Channels, Clustering, and Single-Amino Acid Control of Blood Pressure and Blood Flow.
- Author
-
Jensen LJ
- Subjects
- Animals, Humans, Amino Acids, Calcium Channels metabolism, Hypertension physiopathology, Hypertension drug therapy, Regional Blood Flow, Blood Pressure drug effects, Blood Pressure physiology
- Published
- 2024
- Full Text
- View/download PDF
49. Enhanced isradipine sensitivity in vascular smooth muscle cells due to hypoxia-induced Ca v 1.2 splicing and RbFox1/Fox2 downregulation.
- Author
-
Poore CP, Yang J, Wei S, Fhu CK, Bichler Z, Wang J, Soong TW, and Liao P
- Subjects
- Animals, Rats, Cell Hypoxia genetics, Exons genetics, Mice, Calcium Channel Blockers pharmacology, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Calcium Channels, L-Type metabolism, Calcium Channels, L-Type genetics, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular cytology, RNA Splicing Factors genetics, RNA Splicing Factors metabolism, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle drug effects, Alternative Splicing, Down-Regulation
- Abstract
Calcium influx via the L-type voltage-gated Ca
v 1.2 calcium channel in smooth muscle cells regulates vascular contraction. Calcium channel blockers (CCBs) are widely used to treat hypertension by inhibiting Cav 1.2 channels. Using the vascular smooth muscle cell line, A7r5 and primary culture of cerebral vascular smooth muscle cells, we found that the expression and function of Cav 1.2 channels are downregulated during hypoxia. Furthermore, hypoxia induces structural changes in Cav 1.2 channels via alternative splicing. The expression of exon 9* is upregulated, whereas exon 33 is downregulated. Such structural alterations of Cav 1.2 channels are caused by the decreased expression of RNA-binding proteins RNA-binding protein fox-1 homolog 1 and 2 (RbFox1 and RbFox2). Overexpression of RbFox1 and RbFox2 prevents hypoxia-induced exon 9* inclusion and exon 33 exclusion. Importantly, such structural alterations of the Cav 1.2 channel partly contribute to the enhanced sensitivity of Cav 1.2 to isradipine (a CCB) under hypoxia. Overexpression of RbFox1 and RbFox2 successfully reduces isradipine sensitivity in hypoxic smooth muscle cells. Our results suggest a new strategy to manage ischemic diseases such as stroke and myocardial infarction., (© 2024 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2024
- Full Text
- View/download PDF
50. Post-Translational Modification of Cav1.2 and its Role in Neurodegenerative Diseases
- Author
-
Yun Li, Hong Yang, Tianhan He, Liang Zhang, and Chao Liu
- Subjects
Cav1.2 ,PTM (post-translational modification) ,neurodegenerative disease ,phosphorylation ,ubiquitination ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Cav1.2 plays an essential role in learning and memory, drug addiction, and neuronal development. Intracellular calcium homeostasis is disrupted in neurodegenerative diseases because of abnormal Cav1.2 channel activity and modification of downstream Ca2+ signaling pathways. Multiple post-translational modifications of Cav1.2 have been observed and seem to be closely related to the pathogenesis of neurodegenerative diseases. The specific molecular mechanisms by which Cav1.2 channel activity is regulated remain incompletely understood. Dihydropyridines (DHPs), which are commonly used for hypertension and myocardial ischemia, have been repurposed to treat PD and AD and show protective effects. However, further studies are needed to improve delivery strategies and drug selectivity. Better knowledge of channel modulation and more specific methods for altering Cav1.2 channel function may lead to better therapeutic strategies for neurodegenerative diseases.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.