1
|
Saloman JL, Epouhe AY, Ruff CF, Albers KM. PDX1, a transcription factor essential for organ differentiation, regulates SERCA-dependent Ca 2+ homeostasis in sensory neurons. Cell Calcium 2024; 120:102884. [PMID: 38574509 PMCID: PMC11188734 DOI: 10.1016/j.ceca.2024.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic and duodenal homeobox 1 (PDX1) is a transcription factor required for the development and differentiation of the pancreas. Previous studies indicated that PDX1 expression was restricted to the gastrointestinal tract. Using a cre-dependent reporter, we observed PDX1-dependent expression of tdtomato (PDX1-tom) in a subpopulation of sensory nerves. Many of these PDX1-tom afferents expressed the neurofilament 200 protein and projected to the skin. Tdtomato-labeled terminals were associated with hair follicles in the form of longitudinal and circumferential lanceolate endings suggesting a role in tactile and proprioceptive perception. To begin to examine the functional significance of PDX1 in afferents, we used Fura-2 imaging to examine calcium (Ca2+) handling under naïve and nerve injury conditions. Neuropathic injury is associated with increased intracellular Ca2+ signaling that in part results from dysregulation of the sarco/endoplasmic reticulum calcium transport ATPase (SERCA). Here we demonstrate that under naïve conditions, PDX1 regulates expression of the SERCA2B isoform in sensory neurons. In response to infraorbital nerve injury, a significant reduction of PDX1 and SERCA2B expression and dysregulation of Ca2+ handling occurs in PDX1-tom trigeminal ganglia neurons. The identification of PDX1 expression in the somatosensory system and its regulation of SERCA2B and Ca2+ handling provide a new mechanism to explain pathological changes in primary afferents that may contribute to pain associated with nerve injury.
Collapse
Affiliation(s)
- Jami L Saloman
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Ariel Y Epouhe
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Catherine F Ruff
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Kakizawa S, Kishimoto Y, Yamamoto S, Onga K, Yasuda K, Miyamoto Y, Watanabe M, Sakai R, Mori N. Functional maintenance of calcium store by ShcB adaptor protein in cerebellar Purkinje cells. Sci Rep 2020; 10:14475. [PMID: 32879382 PMCID: PMC7468156 DOI: 10.1038/s41598-020-71414-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/14/2020] [Indexed: 11/15/2022] Open
Abstract
Intracellular Ca2+ levels are changed by influx from extracellular medium and release from intracellular stores. In the central nervous systems, Ca2+ release is involved in various physiological events, such as neuronal excitability and transmitter release. Although stable Ca2+ release in response to stimulus is critical for proper functions of the nervous systems, regulatory mechanisms relating to Ca2+ release are not fully understood in central neurons. Here, we demonstrate that ShcB, an adaptor protein expressed in central neurons, has an essential role in functional maintenance of Ca2+ store in cerebellar Purkinje cells (PCs). ShcB-knockout (KO) mice showed defects in cerebellar-dependent motor function and long-term depression (LTD) at cerebellar synapse. The reduced LTD was accompanied with an impairment of intracellular Ca2+ release. Although the expression of Ca2+ release channels and morphology of Ca2+ store looked intact, content of intracellular Ca2+ store and activity of sarco/endoplasmic reticular Ca2+-ATPase (SERCA) were largely decreased in the ShcB-deficient cerebellum. Furthermore, when ShcB was ectopically expressed in the ShcB-KO PCs, the Ca2+ release and its SERCA-dependent component were restored. These data indicate that ShcB plays a key role in the functional maintenance of ER Ca2+ store in central neurons through regulation of SERCA activity.
Collapse
Affiliation(s)
- Sho Kakizawa
- Department of Anatomy and Neurobiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8523, Japan. .,Department of Biological Chemistry, Graduate School of Pharmaceutical Science, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Yasushi Kishimoto
- Department of Biophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, 769-2193, Japan
| | - Shinichiro Yamamoto
- Department of Biological Chemistry, Graduate School of Pharmaceutical Science, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan.,Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano-ku, Tokyo, 164-8530, Japan
| | - Kazuko Onga
- Department of Anatomy and Neurobiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Kunihiko Yasuda
- Department of Anatomy and Neurobiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8523, Japan.,Department of Occupational Therapy, Faculty of Fukuoka Medical Technology, Teikyo University, Omuta, 836-8505, Japan
| | - Yoshiaki Miyamoto
- Department of Pharmaceutical Therapy and Neuropharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, 060-8638, Japan
| | - Ryuichi Sakai
- Department of Biochemistry, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0373, Japan
| | - Nozomu Mori
- Department of Anatomy and Neurobiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8523, Japan. .,Faculty of Medicine, Fukuoka International University of Health and Welfare, Fukuoka, 814-0001, Japan.
| |
Collapse
|
3
|
Mahajan G, Nadkarni S. Intracellular calcium stores mediate metaplasticity at hippocampal dendritic spines. J Physiol 2019; 597:3473-3502. [PMID: 31099020 PMCID: PMC6636706 DOI: 10.1113/jp277726] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Key points Calcium (Ca2+) entry mediated by NMDA receptors is considered central to the induction of activity‐dependent synaptic plasticity in hippocampal area CA1; this description does not, however, take into account the potential contribution of endoplasmic reticulum (ER) Ca2+ stores. The ER has a heterogeneous distribution in CA1 dendritic spines, and may introduce localized functional differences in Ca2+ signalling between synapses, as suggested by experiments on metabotropic receptor‐dependent long‐term depression. A physiologically detailed computational model of Ca2+ dynamics at a CA3–CA1 excitatory synapse characterizes the contribution of spine ER via metabotropic signalling during plasticity induction protocols. ER Ca2+ release via IP3 receptors modulates NMDA receptor‐dependent plasticity in a graded manner, to selectively promote synaptic depression with relatively diminished effect on LTP induction; this may temper further strengthening at the stronger synapses which are preferentially associated with ER‐containing spines. Acquisition of spine ER may thus represent a local, biophysically plausible ‘metaplastic switch’ at potentiated CA1 synapses, contributing to the plasticity–stability balance in neural circuits.
Abstract Long‐term plasticity mediated by NMDA receptors supports input‐specific, Hebbian forms of learning at excitatory CA3–CA1 connections in the hippocampus. There exists an additional layer of stabilizing mechanisms that act globally as well as locally over multiple time scales to ensure that plasticity occurs in a constrained manner. Here, we investigated the role of calcium (Ca2+) stores associated with the endoplasmic reticulum (ER) in the local regulation of plasticity at individual CA1 synapses. Our study was spurred by (1) the curious observation that ER is sparsely distributed in dendritic spines, but over‐represented in larger spines that are likely to have undergone activity‐dependent strengthening, and (2) evidence suggesting that ER motility at synapses can be rapid, and accompany activity‐regulated spine remodelling. We constructed a physiologically realistic computational model of an ER‐bearing CA1 spine, and examined how IP3‐sensitive Ca2+ stores affect spine Ca2+ dynamics during activity patterns mimicking the induction of long‐term potentiation and long‐term depression (LTD). Our results suggest that the presence of ER modulates NMDA receptor‐dependent plasticity in a graded manner that selectively enhances LTD induction. We propose that ER may locally tune Ca2+‐based plasticity, providing a braking mechanism to mitigate runaway strengthening at potentiated synapses. Our study provides a biophysically accurate description of postsynaptic Ca2+ regulation, and suggests that ER in the spine may promote the re‐use of hippocampal synapses with saturated strengths. Calcium (Ca2+) entry mediated by NMDA receptors is considered central to the induction of activity‐dependent synaptic plasticity in hippocampal area CA1; this description does not, however, take into account the potential contribution of endoplasmic reticulum (ER) Ca2+ stores. The ER has a heterogeneous distribution in CA1 dendritic spines, and may introduce localized functional differences in Ca2+ signalling between synapses, as suggested by experiments on metabotropic receptor‐dependent long‐term depression. A physiologically detailed computational model of Ca2+ dynamics at a CA3–CA1 excitatory synapse characterizes the contribution of spine ER via metabotropic signalling during plasticity induction protocols. ER Ca2+ release via IP3 receptors modulates NMDA receptor‐dependent plasticity in a graded manner, to selectively promote synaptic depression with relatively diminished effect on LTP induction; this may temper further strengthening at the stronger synapses which are preferentially associated with ER‐containing spines. Acquisition of spine ER may thus represent a local, biophysically plausible ‘metaplastic switch’ at potentiated CA1 synapses, contributing to the plasticity–stability balance in neural circuits.
Collapse
Affiliation(s)
- Gaurang Mahajan
- Indian Institute of Science Education and Research, Pune, 411 008, India
| | - Suhita Nadkarni
- Indian Institute of Science Education and Research, Pune, 411 008, India
| |
Collapse
|
4
|
Intravital Two-photon Imaging of Ca 2+ signaling in Secretory Organs of Yellow Cameleon Transgenic Mice. Sci Rep 2018; 8:15880. [PMID: 30367106 PMCID: PMC6203801 DOI: 10.1038/s41598-018-34347-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/06/2018] [Indexed: 01/01/2023] Open
Abstract
Intracellular calcium ([Ca2+]i) signaling regulates physiological functions in most cells. In secretory organs, such as the pancreas, salivary gland, and lacrimal gland (LG), [Ca2+]i elevation in acinar cells triggers fluid secretion, which plays vital roles in the maintenance of functional health across the life-course. It is important to understand the secretory mechanism of secretory organs, but lack of analytic systems available for living animals limits the scope of research to gain deeper insights into the precise mechanism of secretion. We established an intravital imaging system for specific cell types of secretory organs to monitor the [Ca2+]i changes using mouse line expressing Yellow Cameleon 3.60, a genetically encoded Ca2+ indicator. Elevation of [Ca2+]i in specific cell types of secretory organs could be monitored after cholinergic stimulation ex vivo and intravitally. We found that a marked attenuation of LG [Ca2+]i response to cholinergic stimulation was induced under pathological conditions by postganglionic denervation. Intravital Ca2+ imaging in secretory organs will broaden our understanding of the cellular mechanisms in animal models of secretory diseases.
Collapse
|
5
|
Bower DV, Lansdale N, Navarro S, Truong TV, Bower DJ, Featherstone NC, Connell MG, Al Alam D, Frey MR, Trinh LA, Fernandez GE, Warburton D, Fraser SE, Bennett D, Jesudason EC. SERCA directs cell migration and branching across species and germ layers. Biol Open 2017; 6:1458-1471. [PMID: 28821490 PMCID: PMC5665464 DOI: 10.1242/bio.026039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/14/2017] [Indexed: 12/24/2022] Open
Abstract
Branching morphogenesis underlies organogenesis in vertebrates and invertebrates, yet is incompletely understood. Here, we show that the sarco-endoplasmic reticulum Ca2+ reuptake pump (SERCA) directs budding across germ layers and species. Clonal knockdown demonstrated a cell-autonomous role for SERCA in Drosophila air sac budding. Live imaging of Drosophila tracheogenesis revealed elevated Ca2+ levels in migratory tip cells as they form branches. SERCA blockade abolished this Ca2+ differential, aborting both cell migration and new branching. Activating protein kinase C (PKC) rescued Ca2+ in tip cells and restored cell migration and branching. Likewise, inhibiting SERCA abolished mammalian epithelial budding, PKC activation rescued budding, while morphogens did not. Mesoderm (zebrafish angiogenesis) and ectoderm (Drosophila nervous system) behaved similarly, suggesting a conserved requirement for cell-autonomous Ca2+ signaling, established by SERCA, in iterative budding.
Collapse
Affiliation(s)
- Danielle V Bower
- Division of Biological Sciences, California Institute of Technology, Pasadena, CA 91125, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland, and the Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Nick Lansdale
- Department of Biochemistry & Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
- Division of Child Health, Institute of Translational Medicine, University of Liverpool, Liverpool L12 2AP, UK
| | - Sonia Navarro
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Craniofacial Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Thai V Truong
- Division of Biological Sciences, California Institute of Technology, Pasadena, CA 91125, USA
- Biological Sciences and Molecular and Computational Biology, Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Dan J Bower
- Center for Space and Habitability, University of Bern, 3012 Bern, Switzerland
| | - Neil C Featherstone
- Department of Biochemistry & Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Marilyn G Connell
- Department of Biochemistry & Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Denise Al Alam
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Mark R Frey
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Le A Trinh
- Division of Biological Sciences, California Institute of Technology, Pasadena, CA 91125, USA
- Biological Sciences and Molecular and Computational Biology, Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
| | - G Esteban Fernandez
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - David Warburton
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Scott E Fraser
- Division of Biological Sciences, California Institute of Technology, Pasadena, CA 91125, USA
- Biological Sciences and Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Biological Sciences and Molecular and Computational Biology, Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Daimark Bennett
- Department of Biochemistry & Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Edwin C Jesudason
- Division of Biological Sciences, California Institute of Technology, Pasadena, CA 91125, USA
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- NHS Lothian, Edinburgh, EH14 1TY, UK
| |
Collapse
|
6
|
Orai3 channel is the 2-APB-induced endoplasmic reticulum calcium leak. Cell Calcium 2017; 65:91-101. [DOI: 10.1016/j.ceca.2017.01.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 12/22/2022]
|
7
|
Donnarumma E, Trivedi RK, Lefer DJ. Protective Actions of H2S in Acute Myocardial Infarction and Heart Failure. Compr Physiol 2017; 7:583-602. [PMID: 28333381 DOI: 10.1002/cphy.c160023] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hydrogen sulfide (H2S) was identified as the third gasotransmitter in 1996 following the discoveries of the biological importance of nitric oxide and carbon monoxide. Although H2S has long been considered a highly toxic gas, the discovery of its presence and enzymatic production in mammalian tissues supports a critical role for this physiological signaling molecule. H2S is synthesized endogenously by three enzymes: cystathionine β-synthase, cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase. H2S plays a pivotal role in the regulation of cardiovascular function as H2S has been shown to modulate: vasodilation, angiogenesis, inflammation, oxidative stress, and apoptosis. Perturbation of endogenous production of H2S has been associated with many pathological conditions of the cardiovascular system such as diabetes, heart failure, and hypertension. As such, modulation of the endogenous H2S signaling pathway or administration of exogenous H2S has been shown to be cytoprotective. This review article will provide a summary of the current body of evidence on the role of H2S signaling in the setting of myocardial ischemia and heart failure. © 2017 American Physiological Society. Compr Physiol 7:583-602, 2017.
Collapse
Affiliation(s)
- Erminia Donnarumma
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Rishi K Trivedi
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - David J Lefer
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
8
|
Guo Y, Zhang Z, Wu HE, Luo ZD, Hogan QH, Pan B. Increased thrombospondin-4 after nerve injury mediates disruption of intracellular calcium signaling in primary sensory neurons. Neuropharmacology 2017; 117:292-304. [PMID: 28232180 DOI: 10.1016/j.neuropharm.2017.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/31/2017] [Accepted: 02/18/2017] [Indexed: 12/14/2022]
Abstract
Painful nerve injury disrupts Ca2+ signaling in primary sensory neurons by elevating plasma membrane Ca2+-ATPase (PMCA) function and depressing sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) function, which decreases endoplasmic reticulum (ER) Ca2+ stores and stimulates store-operated Ca2+ entry (SOCE). The extracellular matrix glycoprotein thrombospondin-4 (TSP4), which is increased after painful nerve injury, decreases Ca2+ current (ICa) through high-voltage-activated Ca2+ channels and increases ICa through low-voltage-activated Ca2+ channels in dorsal root ganglion neurons, which are events similar to the effect of nerve injury. We therefore examined whether TSP4 plays a critical role in injury-induced disruption of intracellular Ca2+ signaling. We found that TSP4 increases PMCA activity, inhibits SERCA, depletes ER Ca2+ stores, and enhances store-operated Ca2+ influx. Injury-induced changes of SERCA and PMCA function are attenuated in TSP4 knock-out mice. Effects of TSP4 on intracellular Ca2+ signaling are attenuated in voltage-gated Ca2+ channel α2δ1 subunit (Cavα2δ1) conditional knock-out mice and are also Protein Kinase C (PKC) signaling dependent. These findings suggest that TSP4 elevation may contribute to the pathogenesis of chronic pain following nerve injury by disrupting intracellular Ca2+ signaling via interacting with the Cavα2δ1 and the subsequent PKC signaling pathway. Controlling TSP4 mediated intracellular Ca2+ signaling in peripheral sensory neurons may be a target for analgesic drug development for neuropathic pain.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Zhiyong Zhang
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Z David Luo
- Department of Anesthesiology & Perioperative Care, University of California Irvine, Irvine, CA 92697, United States; Department of Pharmacology, University of California Irvine, Irvine, CA 92697, United States
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| |
Collapse
|
9
|
Differential regulation of the Na +-Ca 2+ exchanger 3 (NCX3) by protein kinase PKC and PKA. Cell Calcium 2017; 65:52-62. [PMID: 28233567 DOI: 10.1016/j.ceca.2017.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 11/21/2022]
Abstract
Isoform 3 of the Na+-Ca2+ exchanger (NCX3) participates in the Ca2+ fluxes across the plasma membrane. Among the NCX family, NCX3 carries out a peculiar role due to its specific functions in skeletal muscle and the immune system and to its neuroprotective effect under stress exposure. In this context, proper understanding of the regulation of NCX3 is primordial to consider its potential use as a drug target. In this study, we demonstrated the regulation of NCX3 by protein kinase A (PKA) and C (PKC). Disparity in regulation has been previously reported among the splice variants of NCX3 therefore the activity of Ca2+ uptake and extrusion of the two murine variants was measured using fura-2-based Ca2+ imaging and revealed that both variants are similarly regulated. PKC stimulation diminished the Ca2+ uptake performed by NCX3 in the reverse mode, triggered by a rise in [Ca2+]i or [Na+]i, whereas an opposite response was observed upon PKA stimulation, with a significant increase of the Ca2+ uptake after a rise in [Ca2+]i. The latter stimulation affected similarly the efflux capacity of NCX3 whereas Ca2+ extrusion capacity remained unaffected under activation of PKC. Next, using site-directed mutagenesis, the sensitivity of NCX3 to PKC was abolished by singly mutating its predicted phosphorylation sites T529 or S695. The sensitivity to PKC might be due to the influence of T529 phosphorylation on the Ca2+-binding domain 1. Additionally, we showed that stimulation of NCX3 by PKA occurred through residue S524. This effect may well participate in the fight-or-flight response in skeletal muscle and the long-term potentiation in hippocampus.
Collapse
|
10
|
Rouwette T, Sondermann J, Avenali L, Gomez-Varela D, Schmidt M. Standardized Profiling of The Membrane-Enriched Proteome of Mouse Dorsal Root Ganglia (DRG) Provides Novel Insights Into Chronic Pain. Mol Cell Proteomics 2016; 15:2152-68. [PMID: 27103637 DOI: 10.1074/mcp.m116.058966] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 01/08/2023] Open
Abstract
Chronic pain is a complex disease with limited treatment options. Several profiling efforts have been employed with the aim to dissect its molecular underpinnings. However, generated results are often inconsistent and nonoverlapping, which is largely because of inherent technical constraints. Emerging data-independent acquisition (DIA)-mass spectrometry (MS) has the potential to provide unbiased, reproducible and quantitative proteome maps - a prerequisite for standardization among experiments. Here, we designed a DIA-based proteomics workflow to profile changes in the abundance of dorsal root ganglia (DRG) proteins in two mouse models of chronic pain, inflammatory and neuropathic. We generated a DRG-specific spectral library containing 3067 DRG proteins, which enables their standardized quantification by means of DIA-MS in any laboratory. Using this resource, we profiled 2526 DRG proteins in each biological replicate of both chronic pain models and respective controls with unprecedented reproducibility. We detected numerous differentially regulated proteins, the majority of which exhibited pain model-specificity. Our approach recapitulates known biology and discovers dozens of proteins that have not been characterized in the somatosensory system before. Functional validation experiments and analysis of mouse pain behaviors demonstrate that indeed meaningful protein alterations were discovered. These results illustrate how the application of DIA-MS can open new avenues to achieve the long-awaited standardization in the molecular dissection of pathologies of the somatosensory system. Therefore, our findings provide a valuable framework to qualitatively extend our understanding of chronic pain and somatosensation.
Collapse
Affiliation(s)
- Tom Rouwette
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - Julia Sondermann
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - Luca Avenali
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - David Gomez-Varela
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| | - Manuela Schmidt
- From the ‡Somatosensory Signaling and Systems Biology Group, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Goettingen, Germany
| |
Collapse
|
11
|
Ghosh B, Green MV, Krogh KA, Thayer SA. Interleukin-1β activates an Src family kinase to stimulate the plasma membrane Ca2+ pump in hippocampal neurons. J Neurophysiol 2016; 115:1875-85. [PMID: 26843596 PMCID: PMC4869483 DOI: 10.1152/jn.00541.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 01/23/2016] [Indexed: 01/09/2023] Open
Abstract
The plasma membrane Ca(2+) ATPase (PMCA) plays a major role in clearing Ca(2+) from the neuronal cytoplasm. The cytoplasmic Ca(2+) clearance rate affects neuronal excitability, synaptic plasticity, and neurotransmission. Here, we examined the modulation of PMCA activity by PTKs in hippocampal neurons. PMCA-mediated Ca(2+) clearance slowed in the presence of pyrazolopyrimidine 2, an inhibitor of Src family kinases (SFKs), and accelerated in the presence of C2-ceramide, an activator of PTKs. Ca(2+) clearance kinetics were attenuated in cells expressing a dominant-negative Src mutant, suggesting that the pump is tonically stimulated by a PTK. Tonic stimulation was reduced in hippocampal neurons expressing short hairpin (sh)RNA directed to mRNA for Yes. shRNA-mediated knockdown of PMCA isoform 1 (PMCA1) removed tonic stimulation of Ca(2+) clearance, indicating that the kinase stimulates PMCA1. IL-1β accelerated Ca(2+) clearance in a manner blocked by an IL-1β receptor antagonist or by an inhibitor of neutral sphingomyelinase, the enzyme that produces ceramide. Thus IL-1β activates an SFK to stimulate the plasma membrane Ca(2+) pump, decreasing the duration of Ca(2+) transients in hippocampal neurons.
Collapse
Affiliation(s)
- Biswarup Ghosh
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Matthew V Green
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Kelly A Krogh
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Stanley A Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
12
|
Lever RA, Hussain A, Sun BB, Sage SO, Harper AGS. Conventional protein kinase C isoforms differentially regulate ADP- and thrombin-evoked Ca²⁺ signalling in human platelets. Cell Calcium 2015; 58:577-88. [PMID: 26434503 DOI: 10.1016/j.ceca.2015.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 11/15/2022]
Abstract
Rises in cytosolic Ca(2+) concentration ([Ca(2+)]cyt) are central in platelet activation, yet many aspects of the underlying mechanisms are poorly understood. Most studies examine how experimental manipulations affect agonist-evoked rises in [Ca(2+)]cyt, but these only monitor the net effect of manipulations on the processes controlling [Ca(2+)]cyt (Ca(2+) buffering, sequestration, release, entry and removal), and cannot resolve the source of the Ca(2+) or the transporters or channels affected. To investigate the effects of protein kinase C (PKC) on platelet Ca(2+) signalling, we here monitor Ca(2+) flux around the platelet by measuring net Ca(2+) fluxes to or from the extracellular space and the intracellular Ca(2+) stores, which act as the major sources and sinks for Ca(2+) influx into and efflux from the cytosol, as well as monitoring the cytosolic Na(+) concentration ([Na(+)]cyt), which influences platelet Ca(2+) fluxes via Na(+)/Ca(2+) exchange. The intracellular store Ca(2+) concentration ([Ca(2+)]st) was monitored using Fluo-5N, the extracellular Ca(2+) concentration ([Ca(2+)]ext) was monitored using Fluo-4 whilst [Ca(2+)]cyt and [Na(+)]cyt were monitored using Fura-2 and SFBI, respectively. PKC inhibition using Ro-31-8220 or bisindolylmaleimide I potentiated ADP- and thrombin-evoked rises in [Ca(2+)]cyt in the absence of extracellular Ca(2+). PKC inhibition potentiated ADP-evoked but reduced thrombin-evoked intracellular Ca(2+) release and Ca(2+) removal into the extracellular medium. SERCA inhibition using thapsigargin and 2,5-di(tert-butyl) l,4-benzohydroquinone abolished the effect of PKC inhibitors on ADP-evoked changes in [Ca(2+)]cyt but only reduced the effect on thrombin-evoked responses. Thrombin evokes substantial rises in [Na(+)]cyt which would be expected to reduce Ca(2+) removal via the Na(+)/Ca(2+) exchanger (NCX). Thrombin-evoked rises in [Na(+)]cyt were potentiated by PKC inhibition, an effect which was not due to altered changes in non-selective cation permeability of the plasma membrane as assessed by Mn(2+) quench of Fura-2 fluorescence. PKC inhibition was without effect on thrombin-evoked rises in [Ca(2+)]cyt following SERCA inhibition and either removal of extracellular Na(+) or inhibition of Na(+)/K(+)-ATPase activity by removal of extracellular K(+) or treatment with digoxin. These data suggest that PKC limits ADP-evoked rises in [Ca(2+)]cyt by acceleration of SERCA activity, whilst rises in [Ca(2+)]cyt evoked by the stronger platelet activator thrombin are limited by PKC through acceleration of both SERCA and Na(+)/K(+)-ATPase activity, with the latter limiting the effect of thrombin on rises in [Na(+)]cyt and so forward mode NCX activity. The use of selective PKC inhibitors indicated that conventional and not novel PKC isoforms are responsible for the inhibition of agonist-evoked Ca(2+) signalling.
Collapse
Affiliation(s)
- Robert A Lever
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Azhar Hussain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Benjamin B Sun
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Stewart O Sage
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Alan G S Harper
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom; Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Hartshill, Stoke-on-Trent ST4 7QB, United Kingdom.
| |
Collapse
|
13
|
Morita M, Nakane A, Maekawa S, Kudo Y. Pharmacological characterization of the involvement of protein kinase C in oscillatory and non-oscillatory calcium increases in astrocytes. J Pharmacol Sci 2015; 129:38-42. [PMID: 26349942 DOI: 10.1016/j.jphs.2015.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 08/09/2015] [Accepted: 08/11/2015] [Indexed: 01/24/2023] Open
Abstract
Evidence increasingly shows that astrocytes play a pivotal role in brain physiology and pathology via calcium dependent processes, thus the characterization of the calcium dynamics in astrocytes is of growing importance. We have previously reported that the epidermal growth factor and basic fibroblast growth factor up-regulate the oscillation of the calcium releases that are induced by stimuli, including glutamate in cultured astrocytes. This calcium oscillation is assumed to involve protein kinase C (PKC), which is activated together with the calcium releases as a consequence of inositol phospholipid hydrolysis. In the present study, this issue has been investigated pharmacologically by using astrocytes cultured with and without the growth factors. The pharmacological activation of PKC largely reduced the glutamate-induced oscillatory and non-oscillatory calcium increases. Meanwhile, PKC inhibitors increased the total amounts of both calcium increases without affecting the peak amplitudes and converted the calcium oscillations to non-oscillatory sustained calcium increases by abolishing the falling phases of the repetitive calcium increases. Furthermore, the pharmacological effects were consistent between both glutamate- and histamine-induced calcium oscillations. These results suggest that PKC up-regulates the removal of cytosolic calcium in astrocytes, and this up-regulation is essential for calcium oscillation in astrocytes cultured with growth factors.
Collapse
Affiliation(s)
- Mitsuhiro Morita
- Department of Biology, Kobe University Graduate School of Science, Kobe, 657-8501, Japan; School of Life Science, Tokyo University of Pharmacy and Life Science, Tokyo, 192-0392, Japan.
| | - Akira Nakane
- School of Life Science, Tokyo University of Pharmacy and Life Science, Tokyo, 192-0392, Japan
| | - Shohei Maekawa
- Department of Biology, Kobe University Graduate School of Science, Kobe, 657-8501, Japan
| | - Yoshihisa Kudo
- School of Life Science, Tokyo University of Pharmacy and Life Science, Tokyo, 192-0392, Japan
| |
Collapse
|
14
|
Scheff NN, Yilmaz E, Gold MS. The properties, distribution and function of Na(+)-Ca(2+) exchanger isoforms in rat cutaneous sensory neurons. J Physiol 2014; 592:4969-93. [PMID: 25239455 PMCID: PMC4259538 DOI: 10.1113/jphysiol.2014.278036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/10/2014] [Indexed: 11/08/2022] Open
Abstract
The Na(+)-Ca(2+) exchanger (NCX) appears to play an important role in the regulation of the high K(+)-evoked Ca(2+) transient in putative nociceptive dorsal root ganglion (DRG) neurons. The purpose of the present study was to (1) characterize the properties of NCX activity in subpopulations of DRG neurons, (2) identify the isoform(s) underlying NCX activity, and (3) begin to assess the function of the isoform(s) in vivo. In retrogradely labelled neurons from the glabrous skin of adult male Sprague-Dawley rats, NCX activity, as assessed with fura-2-based microfluorimetry, was only detected in putative nociceptive IB4+ neurons. There were two modes of NCX activity: one was evoked in response to relatively large and long lasting (∼325 nm for >12 s) increases in the concentration of intracellular Ca(2+) ([Ca(2+)]i), and a second was active at resting [Ca(2+)]i > ∼150 nm. There also were two modes of evoked activity: one that decayed relatively rapidly (<5 min) and a second that persisted (>10 min). Whereas mRNA encoding all three NCX isoforms (NCX1-3) was detected in putative nociceptive cutaneous neurons with single cell PCR, pharmacological analysis and small interfering RNA (siRNA) knockdown of each isoform in vivo suggested that NCX2 and 3 were responsible for NCX activity. Western blot analyses suggested that NCX isoforms were differentially distributed within sensory neurons. Functional assays of excitability, action potential propagation, and nociceptive behaviour suggest NCX activity has little influence on excitability per se, but instead influences axonal conduction velocity, resting membrane potential, and nociceptive threshold. Together these results indicate that the function of NCX in the regulation of [Ca(2+)]i in putative nociceptive neurons may be unique relative to other cells in which these exchanger isoforms have been characterized and it has the potential to influence sensory neuron properties at multiple levels.
Collapse
Affiliation(s)
- N N Scheff
- The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - E Yilmaz
- The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - M S Gold
- The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Pan H, Xie X, Chen D, Zhang J, Zhou Y, Yang G. Protective and biogenesis effects of sodium hydrosulfide on brain mitochondria after cardiac arrest and resuscitation. Eur J Pharmacol 2014; 741:74-82. [DOI: 10.1016/j.ejphar.2014.07.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/29/2014] [Accepted: 07/14/2014] [Indexed: 11/26/2022]
|
16
|
Abstract
Hydrogen sulfide (H2S) is an endogenously produced gaseous signaling molecule that elicits a number of cytoprotective effects in mammalian species. H2S was originally considered toxic at elevated levels, but 15 years ago the labile molecule was discovered in mammalian tissue and termed a gasotransmitter, thus opening the door for research aimed towards understanding its physiologic nature. Since then, novel findings have depicted the beneficial aspects of H2S therapy, such as vasodilation, antioxidant upregulation, inflammation inhibition, and activation of anti-apoptotic pathways. These cytoprotective alterations effectively treat multiple forms of cardiac injury at the preclinical level of research. The field has progressed towards instituting novel H2S donors that prove more effective at activating the subsequent cardioprotective enhancements over longer time periods. As more findings explore the efficacy of H2S, research focused on detection of sulfhydrated targets is on the rise. Understanding the molecular mechanisms that stem from H2S treatment may lead the field towards powerful therapeutics in the clinical setting. This review will discuss the cytoprotective and cardioprotective effects of H2S therapy, provide analysis on the molecular alterations that lead to these enhancements, and explore recently developed therapeutics that may bring this gasotransmitter into the clinic in the near future.
Collapse
Affiliation(s)
- Larry A Barr
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine
| | | |
Collapse
|
17
|
Rittiner JE, Brings VE, Zylka MJ. Overexpression of diacylglycerol kinase η enhances Gαq-coupled G protein-coupled receptor signaling. Mol Pharmacol 2014; 85:800-10. [PMID: 24608858 PMCID: PMC3990018 DOI: 10.1124/mol.113.091280] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/07/2014] [Indexed: 11/22/2022] Open
Abstract
Multiple genome-wide association studies have linked diacylglycerol kinase η (DGKη) to bipolar disorder (BPD). Moreover, DGKη expression is increased in tissue from patients with BPD. How increased levels of this lipid kinase might affect cellular functions is currently unclear. Here, we overexpressed mouse DGKη in human embryonic kidney 293 cells to examine substrate specificity and signaling downstream of endogenous G protein-coupled receptors (GPCRs). We found that DGKη can phosphorylate diacylglycerol (DAG) with different acyl side chains (8:0, 12:0, 18:1). In addition, overexpression of DGKη enhanced calcium mobilization after stimulating muscarinic receptors with carbachol and after stimulating purinergic receptors with ATP. This effect required DGKη catalytic activity, as assessed using a kinase-dead (G389D) mutant and multiple truncation constructs. DGKη was localized throughout the cytosol and did not translocate to the plasma membrane after stimulation with carbachol. Since protein kinase C (PKC) can be activated by DAG and promotes receptor desensitization, we also examined functional interactions between PKC and DGKη. We found that acute activation of PKC with phorbol 12-myristate 13-acetate shortened carbachol-evoked calcium responses and occluded the effect of overexpressed DGKη. Moreover, inhibition of PKC activity with bisindolylmaleimide I (BIM I) produced the same enhancing effect on carbachol-evoked calcium mobilization as overexpressed DGKη, and overexpression of DGKη produced no additional effect on calcium mobilization in the presence of BIM I. Taken together, our data suggest that DGKη enhances GPCR signaling by reducing PKC activation.
Collapse
Affiliation(s)
- Joseph E Rittiner
- Department of Cell Biology and Physiology, University of North Carolina Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina
| | | | | |
Collapse
|
18
|
Canonical transient receptor potential channel 2 (TRPC2): old name-new games. Importance in regulating of rat thyroid cell physiology. Pflugers Arch 2014; 466:2025-34. [PMID: 24722829 DOI: 10.1007/s00424-014-1509-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 03/25/2014] [Accepted: 03/26/2014] [Indexed: 12/14/2022]
Abstract
In addition to the TSH-cyclic AMP signalling pathway, calcium signalling is of crucial importance in thyroid cells. Although the importance of calcium signalling has been thoroughly investigated for several decades, the nature of the calcium channels involved in signalling is unknown. In a recent series of investigations using the well-studied rat thyroid FRTL-5 cell line, we showed that these cells exclusively express the transient receptor potential canonical 2 (TRPC2) channel. Our results suggested that the TRPC2 channel is of significant importance in regulating thyroid cell function. These investigations were the first to show that thyroid cells express a member of the TRPC family of ion channels. In this review, we will describe the importance of the TRPC2 channel in regulating TSH receptor expression, thyroglobulin maturation, intracellular calcium and iodide homeostasis and that the channel also regulates thyroid cell proliferation.
Collapse
|
19
|
Liu JX, Hu M, Chen XL, Xu JH, Yang PB, Zhang JS, Liu Y. Reduced expression of Phospholipase C beta in hippocampal interneuron during pilocarpine induced status epilepticus in mice. Neurochem Int 2014; 68:10-7. [PMID: 24480781 DOI: 10.1016/j.neuint.2014.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/02/2014] [Accepted: 01/20/2014] [Indexed: 01/07/2023]
Abstract
We investigated localization of Phospholipase C beta (PLCβ1 and PLCβ4) in laminaes of dorsal hippocampus and different subtypes of hippocampal interneurons in normal Kunming mouse, and their progressive changes during pilocarpine induced status epilepticus (SE) by quantitative immunohistochemistry and real time PCR. PLCβ1 was observed in the pyramidal layer of CA1-3 area, hilus of the dentate gyrus, whereas PLCβ4 was mainly expressed in calcium binding protein positive interneurons, i.e. calbindin, calretinin, parvalbumin positive interneurons in the strata oriens, radiatum of the CA area and hilus of the dentate gyrus. During pilocarpine induced SE, a temporary down-regulation of PLCβ4 in the interneurons of CA area at SE 30min, and a progressive reduction of PLCβ1/PLCβ4 in dentate hilar cells were demonstrated. These findings confirm and extend the regional specific distribution of PLCβ1 and PLCβ4 immunoreactivity in mouse hippocampus, and suggest that PLCβ1 and PLCβ4 may play an important role in maintenance of the status epilepticus.
Collapse
Affiliation(s)
- Jian Xin Liu
- Institute of Neurobiology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Ming Hu
- Institute of Neurobiology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Xin Lin Chen
- Institute of Neurobiology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Jie Hua Xu
- Department of Anatomy and Histology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Peng Bo Yang
- Department of Anatomy and Histology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Jian Shui Zhang
- Institute of Neurobiology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China
| | - Yong Liu
- Institute of Neurobiology, School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, PR China.
| |
Collapse
|
20
|
Scheff NN, Lu SG, Gold MS. Contribution of endoplasmic reticulum Ca2+ regulatory mechanisms to the inflammation-induced increase in the evoked Ca2+ transient in rat cutaneous dorsal root ganglion neurons. Cell Calcium 2013; 54:46-56. [PMID: 23642703 DOI: 10.1016/j.ceca.2013.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/02/2013] [Accepted: 04/04/2013] [Indexed: 12/21/2022]
Abstract
Persistent inflammation results in an increase in the magnitude and duration of high K(+)-evoked Ca(2+) transients in putative nociceptive cutaneous dorsal root ganglion (DRG) neurons. The purpose of the present study was to determine whether recruitment of Ca(2+)-induced Ca(2+) release (CICR) contributes to these inflammation-induced changes. Acutely dissociated, retrogradely labeled cutaneous DRG neurons from naïve and complete Freund's adjuvant inflamed adult male Sprague-Dawley rats were studied with ratiometric microfluorimetry. Ryanodine only attenuated the duration but not magnitude of the high K(+)-evoked Ca(2+) transient in neurons from inflamed rats. However, there was no significant impact of inflammation on the potency or efficacy of ryanodine-induced block of the caffeine-evoked Ca(2+) transient, or the impact of sarco-endoplasmic reticulum ATPase (SERCA) inhibition on the high K(+)-evoked Ca(2+) transient. Furthermore, while there was no change in the magnitude, an inflammation-induced increase in the duration of the caffeine-evoked Ca(2+) transient was only observed with a prolonged caffeine application. In contrast to the high K(+)-evoked Ca(2+) transient, there was no evidence of direct mitochondrial involvement or that of the Ca(2+) extrusion mechanism, the Na(+)/Ca(2+) exchanger, on the caffeine-evoked Ca(2+) transient, and block of SERCA only increased the duration of this transient. These results indicate the presence of Ca(2+) regulatory domains in cutaneous nociceptive DRG neurons within which cytosolic Ca(2+) increased via influx and release are highly segregated. Furthermore, our results suggest that changes in neither CICR machinery nor the coupling between Ca(2+) influx and CICR are primarily responsible for the inflammation-induced changes in the evoked Ca(2+) transient.
Collapse
Affiliation(s)
- Nicole N Scheff
- Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
21
|
Duncan C, Mueller S, Simon E, Renger JJ, Uebele VN, Hogan QH, Wu HE. Painful nerve injury decreases sarco-endoplasmic reticulum Ca²⁺-ATPase activity in axotomized sensory neurons. Neuroscience 2012; 231:247-57. [PMID: 23219911 DOI: 10.1016/j.neuroscience.2012.11.055] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 11/28/2012] [Accepted: 11/29/2012] [Indexed: 12/15/2022]
Abstract
The sarco-endoplasmic reticulum Ca(2+)-ATPase (SERCA) is a critical pathway by which sensory neurons sequester cytosolic Ca(2+) and thereby maintain intracellular Ca(2+) homeostasis. We have previously demonstrated decreased intraluminal endoplasmic reticulum Ca(2+) concentration in traumatized sensory neurons. Here we examine SERCA function in dissociated sensory neurons using Fura-2 fluorometry. Blocking SERCA with thapsigargin (1 μM) increased resting [Ca(2+)](c) and prolonged recovery (τ) from transients induced by neuronal activation (elevated bath K(+)), demonstrating SERCA contributes to control of resting [Ca(2+)](c) and recovery from transient [Ca(2+)](c) elevation. To evaluate SERCA in isolation, plasma membrane Ca(2+) ATPase was blocked with pH 8.8 bath solution and mitochondrial buffering was avoided by keeping transients small (≤ 400 nM). Neurons axotomized by spinal nerve ligation (SNL) showed a slowed rate of transient recovery compared to control neurons, representing diminished SERCA function, whereas neighboring non-axotomized neurons from SNL animals were unaffected. Injury did not affect SERCA function in large neurons. Repeated depolarization prolonged transient recovery, showing that neuronal activation inhibits SERCA function. These findings suggest that injury-induced loss of SERCA function in small sensory neurons may contribute to the generation of pain following peripheral nerve injury.
Collapse
Affiliation(s)
- C Duncan
- Medical College of Wisconsin, Department of Anesthesiology, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Peroxisome proliferator-activated receptor α mediates acute effects of palmitoylethanolamide on sensory neurons. J Neurosci 2012; 32:12735-43. [PMID: 22972997 DOI: 10.1523/jneurosci.0130-12.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The amplitude of the depolarization-evoked Ca2+ transient is larger in dorsal root ganglion (DRG) neurons from tumor-bearing mice compared with that of neurons from naive mice, and the change is mimicked by coculturing DRG neurons with the fibrosarcoma cells used to generate the tumors (Khasabova et al., 2007). The effect of palmitoylethanolamide (PEA), a ligand for the peroxisome proliferator-activated receptor α (PPARα), was determined on the evoked-Ca2+ transient in the coculture condition. The level of PEA was reduced in DRG cells from tumor-bearing mice as well as those cocultured with fibrosarcoma cells. Pretreatment with PEA, a synthetic PPARα agonist (GW7647), or ARN077, an inhibitor of the enzyme that hydrolyzes PEA, acutely decreased the amplitude of the evoked Ca2+ transient in small DRG neurons cocultured with fibrosarcoma cells. The PPARα antagonist GW6471 blocked the effect of each. In contrast, the PPARα agonist was without effect in the control condition, but the antagonist increased the amplitude of the Ca2+ transient, suggesting that PPARα receptors are saturated by endogenous ligand under basal conditions. Effects of drugs on mechanical sensitivity in vivo paralleled their effects on DRG neurons in vitro. Local injection of ARN077 decreased mechanical hyperalgesia in tumor-bearing mice, and the effect was blocked by GW6471. These data support the conclusion that the activity of DRG neurons is rapidly modulated by PEA through a PPARα-dependent mechanism. Moreover, agents that increase the activity of PPARα may provide a therapeutic strategy to reduce tumor-evoked pain.
Collapse
|
23
|
Hobbs RP, Amargo EV, Somasundaram A, Simpson CL, Prakriya M, Denning MF, Green KJ. The calcium ATPase SERCA2 regulates desmoplakin dynamics and intercellular adhesive strength through modulation of PKCα signaling. FASEB J 2011; 25:990-1001. [PMID: 21156808 PMCID: PMC3042836 DOI: 10.1096/fj.10-163261] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 11/24/2010] [Indexed: 11/11/2022]
Abstract
Darier's disease (DD) is an inherited autosomal-dominant skin disorder characterized histologically by loss of adhesion between keratinocytes. DD is typically caused by mutations in sarcoendoplasmic reticulum Ca(2+)-ATPase isoform 2 (SERCA2), a major regulator of intracellular Ca(2+) homeostasis in the skin. However, a defined role for SERCA2 in regulating intercellular adhesion remains poorly understood. We found that diminution of SERCA2 function by pharmacological inhibition or siRNA silencing in multiple human epidermal-derived cell lines was sufficient to disrupt desmosome assembly and weaken intercellular adhesive strength. Specifically, SERCA2-deficient cells exhibited up to a 60% reduction in border translocation of desmoplakin (DP), the desmosomal cytolinker protein necessary for intermediate filament (IF) anchorage to sites of robust cell-cell adhesion. In addition, loss of SERCA2 impaired the membrane translocation of protein kinase C α (PKCα), a known regulator of DP-IF association and desmosome assembly, to the plasma membrane by up to 70%. Exogenous activation of PKCα in SERCA2-deficient cells was sufficient to rescue the defective DP localization, desmosome assembly, and intercellular adhesive strength to levels comparable to controls. Our findings indicate that SERCA2-deficiency is sufficient to impede desmosome assembly and weaken intercellular adhesive strength via a PKCα-dependent mechanism, implicating SERCA2 as a novel regulator of PKCα signaling.
Collapse
Affiliation(s)
| | | | | | | | - Murali Prakriya
- Department of Molecular Pharmacology and Biological Chemistry
| | - Mitchell F. Denning
- Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, Illinois, USA
| | - Kathleen J. Green
- Department of Pathology
- Department of Dermatology, and
- R. H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; and
| |
Collapse
|
24
|
Ghosh B, Li Y, Thayer SA. Inhibition of the plasma membrane Ca2+ pump by CD44 receptor activation of tyrosine kinases increases the action potential afterhyperpolarization in sensory neurons. J Neurosci 2011; 31:2361-70. [PMID: 21325503 PMCID: PMC3565434 DOI: 10.1523/jneurosci.5764-10.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/06/2010] [Accepted: 12/14/2010] [Indexed: 11/21/2022] Open
Abstract
The cytoplasmic Ca(2+) clearance rate affects neuronal excitability, plasticity, and synaptic transmission. Here, we examined the modulation of the plasma membrane Ca(2+) ATPase (PMCA) by tyrosine kinases. In rat sensory neurons grown in culture, the PMCA was under tonic inhibition by a member of the Src family of tyrosine kinases (SFKs). Ca(2+) clearance accelerated in the presence of selective tyrosine kinase inhibitors. Tonic inhibition of the PMCA was attenuated in cells expressing a dominant-negative construct or shRNA directed to message for the SFKs Lck or Fyn, but not Src. SFKs did not appear to phosphorylate the PMCA directly but instead activated focal adhesion kinase (FAK). Expression of constitutively active FAK enhanced and dominant-negative or shRNA knockdown of FAK attenuated tonic inhibition. Antisense knockdown of PMCA isoform 4 removed tonic inhibition of Ca(2+) clearance, indicating that FAK acts on PMCA4. The hyaluronan receptor CD44 activates SFK-FAK signaling cascades and is expressed in sensory neurons. Treating neurons with a CD44-blocking antibody or short hyaluronan oligosaccharides, which are produced during injury and displace macromolecular hyaluronan from CD44, attenuated tonic PMCA inhibition. Ca(2+)-activated K(+) channels mediate a slow afterhyperpolarization in sensory neurons that was inhibited by tyrosine kinase inhibitors and enhanced by knockdown of PMCA4. Thus, we describe a novel kinase cascade in sensory neurons that enables the extracellular matrix to alter Ca(2+) signals by modulating PMCA-mediated Ca(2+) clearance. This signaling pathway may influence the excitability of sensory neurons following injury.
Collapse
Affiliation(s)
- Biswarup Ghosh
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Yan Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| |
Collapse
|
25
|
Phe27Cys polymorphism of the human delta opioid receptor predisposes cells to compromised calcium signaling. Mol Cell Biochem 2011; 351:173-81. [PMID: 21234650 DOI: 10.1007/s11010-011-0725-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 01/05/2011] [Indexed: 10/18/2022]
Abstract
A quarter of the human population with European background carries at least one allele of the OPRD1 gene that encodes the delta opioid receptor with cysteine at the amino acid position 27 (hδOR(Cys27)) instead of the evolutionary conserved phenylalanine (hδOR(Phe27)). The two variants have indistinguishable pharmacological properties but, importantly, hδOR(Cys27) differs from hδOR(Phe27) in having low maturation efficiency, lower stability at the cell surface and pronounced intracellular location. Both variants were previously shown to interact with the Sarco(endo)plasmic reticulum Ca²+ ATPase (SERCA) 2b in the early phase of their biosynthesis. We analyzed by pulse-chase assays, whether cellular signaling can affect hδOR(Cys27) maturation. Neither activation of the receptor by a δOR-specific agonist Leu-enkephalin, induction of intracellular calcium (Ca²+) release by ATP nor the direct stimulation of SERCA 2b by protein kinase C activation affected receptor maturation in HEK-293 cells. No signaling-mediated regulation of receptor maturation could therefore be demonstrated. Instead, we found by using single cell Ca²+ measurements that over-expression of hδOR(Cys27), but not hδOR(Phe27), compromised ATP-induced intracellular Ca²+-signaling. Furthermore, hδOR(Cys27) precursors showed slower dissociation from SERCA2b and hδOR(Cys27) expression caused down-regulation of the homocysteine-inducible endoplasmic reticulum-resident ubiquitin domain-like member 1 protein (HERP). We suggest that aging individuals with at least one hδOR(Cys27) encoding allele might have lowered threshold for Ca²+ dysregulation in neurons expressing hδOR.
Collapse
|
26
|
Merriam LA, Locknar SA, Girard BM, Parsons RL. Somatic ATP release from guinea pig sympathetic neurons does not require calcium-induced calcium release from internal stores. Am J Physiol Cell Physiol 2010; 299:C836-43. [PMID: 20668213 PMCID: PMC2957269 DOI: 10.1152/ajpcell.00036.2010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 07/22/2010] [Indexed: 11/22/2022]
Abstract
Prior studies indicated that a Ca(2+)-dependent release of ATP can be initiated from the soma of sympathetic neurons dissociated from guinea pig stellate ganglia. Previous studies also indicated that Ca(2+)-induced Ca(2+) release (CICR) can modulate membrane excitability in these same neurons. As Ca(2+) release from internal stores is thought to support somatodendritic transmitter release in other neurons, the present study investigated whether CICR is essential for somatic ATP release from dissociated sympathetic neurons. Caffeine increased intracellular Ca(2+) and activated two inward currents: a slow inward current (SIC) in 85% of cells, and multiple faster inward currents [asynchronous transient inward currents (ASTICs)] in 40% of cells voltage-clamped to negative potentials. Caffeine evoked both currents when cells were bathed in a Ca(2+)-deficient solution, indicating that both were initiated by Ca(2+) release from ryanodine-sensitive stores in the endoplasmic reticulum. Sodium influx contributed to generation of both SICs and ASTICs, but only ASTICs were inhibited by the presence of the P2X receptor blocker PPADs. Thus ASTICs, but not SICs, resulted from an ATP activation of P2X receptors. Ionomycin induced ASTICs in a Ca(2+)-containing solution, but not when it was applied in a Ca(2+)-deficient solution, demonstrating the key requirement for external Ca(2+) in initiating ASTICs by ionomycin. Pretreatment with drugs to deplete the internal stores of Ca(2+) did not block the ability of ionomycin or long depolarizing voltage steps to initiate ASTICs. Although a caffeine-induced release of Ca(2+) from internal stores can elicit both SICs and ASTICs in dissociated sympathetic neurons, CICR is not required for the somatic release of ATP.
Collapse
Affiliation(s)
- Laura A Merriam
- Department of Anatomy and Neurobiology, Univ. of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
27
|
Calvert JW, Coetzee WA, Lefer DJ. Novel insights into hydrogen sulfide--mediated cytoprotection. Antioxid Redox Signal 2010; 12:1203-17. [PMID: 19769484 PMCID: PMC2864658 DOI: 10.1089/ars.2009.2882] [Citation(s) in RCA: 256] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydrogen sulfide (H(2)S) is a colorless, water soluble, flammable gas that has the characteristic smell of rotten eggs. Like other members of the gasotransmitter family (nitric oxide and carbon monoxide), H(2)S has traditionally been considered to be a highly toxic gas and environmental hazard. However, much like for nitric oxide and carbon monoxide, the initial negative perception of H(2)S has evolved with the discovery that H(2)S is produced enzymatically in mammals under normal conditions. As a result of this discovery, there has been a great deal of work to elucidate the physiological role of H(2)S. H(2)S is now recognized to be cytoprotective in various models of cellular injury. Specifically, it has been demonstrated that the acute administration of H(2)S, either prior to ischemia or at reperfusion, significantly ameliorates in vitro or in vivo myocardial and hepatic ischemia-reperfusion injury. These studies have also demonstrated a cardioprotective role for endogenous H(2)S. This review article summarizes the current body of evidence demonstrating the cytoprotective effects of H(2)S with an emphasis on the cardioprotective effects. This review also provides a detailed description of the current signaling mechanisms shown to be responsible for these cardioprotective actions.
Collapse
Affiliation(s)
- John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | |
Collapse
|
28
|
Mitochondrial Ca2+ cycling facilitates activation of the transcription factor NFAT in sensory neurons. J Neurosci 2009; 29:12101-14. [PMID: 19793968 DOI: 10.1523/jneurosci.3384-09.2009] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ca(2+)-dependent gene regulation controls many aspects of neuronal plasticity. Significant progress has been made toward understanding the roles of voltage- and ligand-gated Ca(2+) channels in triggering specific transcriptional responses. In contrast, the functional importance of Ca(2+) buffers and Ca(2+) transporters in neuronal gene regulation is less clear despite their critical contribution to the spatiotemporal control of Ca(2+) signals. Here we examined the role of mitochondrial Ca(2+) uptake and release in regulating the Ca(2+)-dependent transcription factor NFAT (nuclear factor of activated T-cells), which has been implicated in synaptic plasticity, axonal growth, and neuronal survival. Intense stimulation of sensory neurons by action potentials or TRPV1 agonists induced rapid activation and nuclear import of NFAT. Nuclear translocation of NFAT was associated with a characteristic prolonged [Ca(2+)](i) elevation (plateau) that resulted from Ca(2+) uptake by, and its subsequent release from, mitochondria. Measurements using a mitochondrial Ca(2+) indicator, mtPericam, showed that this process recruited mitochondria throughout the cell body, including the perinuclear region. [Ca(2+)](i) levels attained during the plateau phase were similar to or higher than those required for NFAT activation (200-300 nm). The elimination of the [Ca(2+)](i) plateau by blocking either mitochondrial Ca(2+) uptake via the uniporter or Ca(2+) release via the mitochondrial Na(+)/Ca(2+) exchanger strongly reduced nuclear import of NFAT. Furthermore, preventing Ca(2+) mobilization via the mitochondrial Na(+)/Ca(2+) exchanger diminished NFAT-mediated transcription. Collectively, these data implicate activity-induced Ca(2+) uptake and prolonged release from mitochondria as a novel regulatory mechanism in neuronal excitation-transcription coupling.
Collapse
|
29
|
Rigaud M, Gemes G, Weyker PD, Cruikshank JM, Kawano T, Wu HE, Hogan QH. Axotomy depletes intracellular calcium stores in primary sensory neurons. Anesthesiology 2009; 111:381-92. [PMID: 19602958 PMCID: PMC2891519 DOI: 10.1097/aln.0b013e3181ae6212] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The cellular mechanisms of neuropathic pain are inadequately understood. Previous investigations have revealed disrupted Ca signaling in primary sensory neurons after injury. The authors examined the effect of injury on intracellular Ca stores of the endoplasmic reticulum, which critically regulate the Ca signal and neuronal function. METHODS Intracellular Ca levels were measured with Fura-2 or mag-Fura-2 microfluorometry in axotomized fifth lumbar (L5) dorsal root ganglion neurons and adjacent L4 neurons isolated from hyperalgesic rats after L5 spinal nerve ligation, compared to neurons from control animals. RESULTS Endoplasmic reticulum Ca stores released by the ryanodine-receptor agonist caffeine decreased by 46% in axotomized small neurons. This effect persisted in Ca-free bath solution, which removes the contribution of store-operated membrane Ca channels, and after blockade of the mitochondrial, sarco-endoplasmic Ca-ATPase and the plasma membrane Ca ATPase pathways. Ca released by the sarco-endoplasmic Ca-ATPase blocker thapsigargin and by the Ca-ionophore ionomycin was also diminished by 25% and 41%, respectively. In contrast to control neurons, Ca stores in axotomized neurons were not expanded by neuronal activation by K depolarization, and the proportionate rate of refilling by sarco-endoplasmic Ca-ATPase was normal. Luminal Ca concentration was also reduced by 38% in axotomized neurons in permeabilized neurons. The adjacent neurons of the L4 dorsal root ganglia showed modest and inconsistent changes after L5 spinal nerve ligation. CONCLUSIONS Painful nerve injury leads to diminished releasable endoplasmic reticulum Ca stores and a reduced luminal Ca concentration. Depletion of Ca stores may contribute to the pathogenesis of neuropathic pain.
Collapse
Affiliation(s)
- Marcel Rigaud
- Research Fellow, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Resident, Department of Anesthesiology, Medical University of Graz, Graz, Austria
| | - Geza Gemes
- Research Fellow, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Resident, Department of Anesthesiology, Medical University of Graz, Graz, Austria
| | - Paul D. Weyker
- Medical Student, University of Wisconsin, Madison, Wisconsin
| | - James M. Cruikshank
- Research Assistant, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Takashi Kawano
- Research Fellow, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hsiang-En Wu
- Assistant Professor, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Quinn H. Hogan
- Professor, Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Anesthesiologist, Zablocki VA Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
30
|
Ferragamo MJ, Reinardy JL, Thayer SA. Ca2+-dependent, stimulus-specific modulation of the plasma membrane Ca2+ pump in hippocampal neurons. J Neurophysiol 2009; 101:2563-71. [PMID: 19244356 DOI: 10.1152/jn.90774.2008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The plasma membrane Ca(2+) ATPase (PMCA) plays a major role in restoring Ca(2+) to basal levels following transient elevation by neuronal activity. Here we examined the effects of various stimuli that increase [Ca(2+)](i) on PMCA-mediated Ca(2+) clearance from hippocampal neurons. We used indo-1-based microfluorimetry in the presence of cyclopiazonic acid to study the rate of PMCA-mediated recovery of Ca(2+) elevated by a brief train of action potentials. [Ca(2+)](i) recovery was described by an exponential decay and the time constant provided an index of PMCA-mediated Ca(2+) clearance. PMCA function was assessed before and for >or=60 min following a 10-min priming stimulus of either 100 microM N-methyl-d-aspartate (NMDA), 0.1 mM Mg(2+) (reduced extracellular Mg(2+) induces intense excitatory synaptic activity), 30 mM K(+), or control buffer. Recovery kinetics slowed progressively following priming with NMDA or 0.1 mM Mg(2+); in contrast, Ca(2+) clearance initially accelerated and then slowly returned to initial rates following priming with 30 mM K(+)-induced depolarization. Treatment with 10 muM calpeptin, an inhibitor of the Ca(2+) activated protease calpain, prevented the slowing of kinetics observed following treatment with NMDA but had no affect on the recovery kinetics of control cells. Calpeptin also blocked the rapid acceleration of Ca(2+) clearance following depolarization. In calpeptin-treated cells, 0.1 mM Mg(2+) induced a graded acceleration of Ca(2+) clearance. Thus in spite of producing comparable increases in [Ca(2+)](i), activation of NMDA receptors, depolarization-induced activation of voltage-gated Ca(2+) channels and excitatory synaptic activity each uniquely affected Ca(2+) clearance kinetics mediated by the PMCA.
Collapse
Affiliation(s)
- Michael J Ferragamo
- Department of Biology, Gustavus Adolphus College, Saint Peter, Minneapolis, MN, USA
| | | | | |
Collapse
|
31
|
Abstract
The fundamental role of calcium ions (Ca(2+)) in an excitable tissue, the frog heart, was first demonstrated in a series of classical reports by Sydney Ringer in the latter part of the nineteenth century (1882a, b; 1893a, b). Even so, nearly a century elapsed before it was proven that Ca(2+) regulated the excitability of primary sensory neurons. In this chapter we review the sites and mechanisms whereby internal and external Ca(2+) can directly or indirectly alter the excitability of primary sensory neurons: excitability changes being manifested typically by variations in shape of the action potential or the pattern of its discharge.
Collapse
|
32
|
Fuortes MG, Faria LC, Merlin LR. Impact of protein kinase C activation on epileptiform activity in the hippocampal slice. Epilepsy Res 2008; 82:38-45. [PMID: 18715754 PMCID: PMC2633120 DOI: 10.1016/j.eplepsyres.2008.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 06/03/2008] [Accepted: 07/02/2008] [Indexed: 11/24/2022]
Abstract
There is evidence suggesting that protein kinase C (PKC) activation can prevent the enhanced network excitability associated with status epilepticus and group I metabotropic glutamate receptor (mGluR)-induced epileptogenesis. However, we observed no suppression of mGluR-induced burst prolongation in the guinea pig hippocampal slice when applied in the presence of the PKC activator phorbol-12,13-dibutyrate (PDBu). Furthermore, PDBu alone converted picrotoxin-induced interictal bursts into ictal-length discharges ranging from 2 to 6s in length. This effect could not be elicited by the inactive analog 4-alpha-PDBu and was suppressed with the PKC inhibitor chelerythrine, indicating PKC dependence. PKC activation can enhance neurotransmitter release, and both glutamate and acetylcholine are capable of eliciting similar prolonged synchronized discharges. However, neither mGluR1 nor NMDA receptor antagonist suppressed PDBu-driven burst prolongation, suggesting that increased glutamate release alone is unlikely to account for the PKC-induced expression of ictaform discharges. Similarly, atropine, a broad-spectrum muscarinic receptor antagonist, had no effect on PKC-induced burst prolongation. By contrast, AMPA/kainate receptor antagonist abolished PKC-induced burst prolongation, and mGluR5 antagonist significantly blunted the maximum burst length induced by PKC. These data suggest that PKC-induced prolongation of epileptiform bursts is dependent on changes specific to mGluR5 and AMPA/kainate receptors and not mediated simply by a generalized increase in transmitter release.
Collapse
Affiliation(s)
- Michaelangelo G. Fuortes
- Neural and Behavioral Sciences Program, School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| | - Leonardo C. Faria
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| | - Lisa R. Merlin
- Neural and Behavioral Sciences Program, School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| |
Collapse
|
33
|
Geiger JE, Magoski NS. Ca2+-induced Ca2+ release in Aplysia bag cell neurons requires interaction between mitochondrial and endoplasmic reticulum stores. J Neurophysiol 2008; 100:24-37. [PMID: 18463180 DOI: 10.1152/jn.90356.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intracellular Ca2+ is influenced by both Ca2+ influx and release. We examined intracellular Ca2+ following action potential firing in the bag cell neurons of Aplysia californica. Following brief synaptic input, these neuroendocrine cells undergo an afterdischarge, resulting in elevated Ca2+ and the secretion of neuropeptides to initiate reproduction. Cultured bag cell neurons were injected with the Ca2+ indicator, fura-PE3, and subjected to simultaneous imaging and electrophysiology. Delivery of a 5-Hz, 1-min train of action potentials (mimicking the fast phase of the afterdischarge) produced a Ca2+ rise that markedly outlasted the initial influx, consistent with Ca2+-induced Ca2+ release (CICR). This response was attenuated by about half with ryanodine or depletion of the endoplasmic reticulum (ER) by cyclopiazonic acid. However, depletion of the mitochondria, with carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone, essentially eliminated CICR. Dual depletion of the ER and mitochondria did not reduce CICR further than depletion of the mitochondria alone. Moreover, tetraphenylphosphonium, a blocker of mitochondrial Ca2+ release, largely prevented CICR. The Ca2+ elevation during and subsequent to a stimulus mimicking the full afterdischarge was prominent and enhanced by protein kinase C activation. Traditionally, the ER is seen as the primary Ca2+ source for CICR. However, bag cell neuron CICR represents a departure from this view in that it relies on store interaction, where Ca2+ released from the mitochondria may in turn liberate Ca2+ from the ER. This unique form of CICR may be used by both bag cell neurons, and other neurons, to initiate secretion, activate channels, or induce gene expression.
Collapse
Affiliation(s)
- Julia E Geiger
- Department of Physiology, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
34
|
Powell AD, Toescu EC, Collinge J, Jefferys JGR. Alterations in Ca2+-buffering in prion-null mice: association with reduced afterhyperpolarizations in CA1 hippocampal neurons. J Neurosci 2008; 28:3877-86. [PMID: 18400886 PMCID: PMC6670463 DOI: 10.1523/jneurosci.0675-08.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 02/24/2007] [Indexed: 11/21/2022] Open
Abstract
Prion protein (PrP) is a normal component of neurons, which confers susceptibility to prion diseases. Despite its evolutionary conservation, its normal function remains controversial. PrP-deficient (Prnp(0/0)) mice have weaker afterhyperpolarizations (AHPs) in cerebellar and hippocampal neurons. Here we show that the AHP impairment in hippocampal CA1 pyramidal cells is selective for the slow AHP, and is not caused by an impairment of either voltage-gated Ca(2+) channels or Ca(2+)-activated K(+) channels. Instead, Prnp(0/0) neurons have twofold to threefold stronger Ca(2+) buffering and double the Ca(2+) extrusion rate. In Prnp(0/0) neurons thapsigargin abolished the stronger Ca(2+) buffering and extrusion, and thapsigargin or cyclopiazonic acid abolished the weakening of the slow AHPs. These data implicate sarcoplasmic/endoplasmic reticulum calcium ATPase in the enhanced Ca(2+) buffering, and extrusion into the endoplasmic reticulum, which contains substantial amounts of PrP in wild-type mice. Altered Ca(2+) homeostasis can explain several phenotypes identified in Prnp(0/0) mice.
Collapse
Affiliation(s)
| | - Emil C. Toescu
- Medical Sciences (Physiology), The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom, and
| | - John Collinge
- Medical Research Center Prion Unit, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | | |
Collapse
|
35
|
Yu YG, Tang FG, Pan J, Gu XF. Effects of Phenylalanine and its Metabolites on Cytoplasmic Free Calcium in Cortical Neurons. Neurochem Res 2007; 32:1292-301. [PMID: 17401656 DOI: 10.1007/s11064-007-9303-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2006] [Accepted: 01/30/2007] [Indexed: 01/16/2023]
Abstract
Classic phenylketonuria (PKU) is characterized by brain lesions. However, its underlying neurotoxic mechanisms remain unknown. Based on our previous studies, we hypothesized that calcium might participate in PKU-associated neuropathy. In cultured cortical neurons, cytoplasmic free calcium concentration ([Ca(2+)](i)) decreased dramatically when treatment with phenylalanine (Phe) and phenyllactic acid, while phenylacetic acid treatment immediately increased [Ca(2+)](i), which began to decrease after 3 min. Moreover, [Ca(2+)](i) decreased dramatically after Phe treatment in the presence of EGTA suggesting that Phe might increase [Ca(2+)](i) efflux. Phe-induced [Ca(2+)](i) decrease was strongly inhibited by vanadate, a non-specific plasma membrane Ca(2+)-ATPase (PMCA) antagonist, suggesting that Phe might increase [Ca(2+)](i) efflux throught modulating PMCA. These findings were further supported by the facts that Phe could increase membrance (45)Ca-uptake capability and PMCA activity. In contrast, treatment of KBR7943 or thapsigargin, antagonists to Na/Ca Exchanger (NCX) and Sarco/Endoplasmic reticulum Ca(2+)-ATPase (SERCA), respectively, did not elicit any changes in [Ca(2+)](i). Specific siRNA against PMCA had an effect similar to vanadate. Since the brain injury induced by phenylalaninemia was thought to be a chronic process, we cultured cortical neurons in the presence of Phe for 2 weeks and measured [Ca(2+)](i), PMCA activity and (45)Ca-uptake capability at days 3, 7, 9 and 14, respectively. PMCA activity and (45)Ca-uptake capability decreased from day 9, at the same time [Ca(2+)](i) increase was observed. In conclusion, PMCA participate in regulating Phe-induced initial rapid decrease in [Ca(2+)](i) and subsequent long-term increase in [Ca(2+)](i).
Collapse
Affiliation(s)
- Y G Yu
- Department of Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai, China.
| | | | | | | |
Collapse
|
36
|
Jackson JG, Thayer SA. Mitochondrial modulation of Ca2+ -induced Ca2+ -release in rat sensory neurons. J Neurophysiol 2006; 96:1093-104. [PMID: 16760347 DOI: 10.1152/jn.00283.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ca2+ -induced Ca2+ -release (CICR) from ryanodine-sensitive Ca2+ stores provides a mechanism to amplify and propagate a transient increase in intracellular calcium concentration ([Ca2+]i). A subset of rat dorsal root ganglion neurons in culture exhibited regenerative CICR when sensitized by caffeine. [Ca2+]i oscillated in the maintained presence of 5 mM caffeine and 25 mM K+. Here, CICR oscillations were used to study the complex interplay between Ca2+ regulatory mechanisms at the cellular level. Oscillations depended on Ca2+ uptake and release from the endoplasmic reticulum (ER) and Ca2+ influx across the plasma membrane because cyclopiazonic acid, ryanodine, and removal of extracellular Ca2+ terminated oscillations. Increasing caffeine concentration decreased the threshold for action potential-evoked CICR and increased oscillation frequency. Mitochondria regulated CICR by providing ATP and buffering [Ca2+]i. Treatment with the ATP synthase inhibitor, oligomycin B, decreased oscillation frequency. When ATP concentration was held constant by recording in the whole cell patch-clamp configuration, oligomycin no longer affected oscillation frequency. Aerobically derived ATP modulated CICR by regulating the rate of Ca2+ sequestration by the ER Ca2+ pump. Neither CICR threshold nor Ca2+ clearance by the plasma membrane Ca2+ pump were affected by inhibition of aerobic metabolism. Uncoupling electron transport with carbonyl cyanide p-trifluoromethoxy-phenyl-hydrazone or inhibiting mitochondrial Na+/Ca2+ exchange with CGP37157 revealed that mitochondrial buffering of [Ca2+]i slowed oscillation frequency, decreased spike amplitude, and increased spike width. These findings illustrate the interdependence of energy metabolism and Ca2+ signaling that results from the complex interaction between the mitochondrion and the ER in sensory neurons.
Collapse
Affiliation(s)
- Joshua G Jackson
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
| | | |
Collapse
|