1
|
Elmore G, Ahern BM, McVay NM, Barker KW, Lohano SS, Ali N, Sebastian A, Andres DA, Satin J, Levitan BM. The C-terminus of Rad is required for membrane localization and L-type calcium channel regulation. J Gen Physiol 2024; 156:e202313518. [PMID: 38990175 PMCID: PMC11244639 DOI: 10.1085/jgp.202313518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/17/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
L-type CaV1.2 current (ICa,L) links electrical excitation to contraction in cardiac myocytes. ICa,L is tightly regulated to control cardiac output. Rad is a Ras-related, monomeric protein that binds to L-type calcium channel β subunits (CaVβ) to promote inhibition of ICa,L. In addition to CaVβ interaction conferred by the Rad core motif, the highly conserved Rad C-terminus can direct membrane association in vitro and inhibition of ICa,L in immortalized cell lines. In this work, we test the hypothesis that in cardiomyocytes the polybasic C-terminus of Rad confers t-tubular localization, and that membrane targeting is required for Rad-dependent ICa,L regulation. We introduced a 3xFlag epitope to the N-terminus of the endogenous mouse Rrad gene to facilitate analysis of subcellular localization. Full-length 3xFlag-Rad (Flag-Rad) mice were compared with a second transgenic mouse model, in which the extended polybasic C-termini of 3xFlag-Rad was truncated at alanine 277 (Flag-RadΔCT). Ventricular cardiomyocytes were isolated for anti-Flag-Rad immunocytochemistry and ex vivo electrophysiology. Full-length Flag-Rad showed a repeating t-tubular pattern whereas Flag-RadΔCT failed to display membrane association. ICa,L in Flag-RadΔCT cardiomyocytes showed a hyperpolarized activation midpoint and an increase in maximal conductance. Additionally, current decay was faster in Flag-RadΔCT cells. Myocardial ICa,L in a Rad C-terminal deletion model phenocopies ICa,L modulated in response to β-AR stimulation. Mechanistically, the polybasic Rad C-terminus confers CaV1.2 regulation via membrane association. Interfering with Rad membrane association constitutes a specific target for boosting heart function as a treatment for heart failure with reduced ejection fraction.
Collapse
Affiliation(s)
- Garrett Elmore
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Brooke M Ahern
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Nicholas M McVay
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Kyle W Barker
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Sarisha S Lohano
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Nemat Ali
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Andrea Sebastian
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Bryana M Levitan
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- Gill Heart and Vascular Institute , Lexington, KY, USA
| |
Collapse
|
2
|
Spooner HC, Costa AD, González AH, Ibrahimkhail H, Yarov-Yarovoy V, Horne M, Dickson EJ, Dixon RE. 14-3-3 promotes sarcolemmal expression of cardiac Ca V 1.2 and nucleates isoproterenol-triggered channel super-clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.607987. [PMID: 39229175 PMCID: PMC11370440 DOI: 10.1101/2024.08.16.607987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The L-type Ca 2+ channel (Ca V 1.2) is essential for cardiac excitation-contraction coupling. To contribute to the inward Ca 2+ flux that drives Ca 2+ -induced-Ca 2+ -release, Ca V 1.2 channels must be expressed on the sarcolemma; thus the regulatory mechanisms that tune Ca V 1.2 expression to meet contractile demand are an emerging area of research. A ubiquitously expressed protein called 14-3-3 has been proposed to affect Ca 2+ channel trafficking in non-myocytes, however whether 14-3-3 has similar effects on Ca V 1.2 in cardiomyocytes is unknown. 14-3-3 preferentially binds phospho-serine/threonine residues to affect many cellular processes and is known to regulate cardiac ion channels including Na V 1.5 and hERG. Altered 14-3-3 expression and function have been implicated in cardiac pathologies including hypertrophy. Accordingly, we tested the hypothesis that 14-3-3 interacts with Ca V 1.2 in a phosphorylation-dependent manner and regulates cardiac Ca V 1.2 trafficking and recycling. Confocal imaging, proximity ligation assays, super-resolution imaging, and co-immunoprecipitation revealed a population of 14-3-3 colocalized and closely associated with Ca V 1.2. The degree of 14-3-3/Ca V 1.2 colocalization increased upon stimulation of β -adrenergic receptors with isoproterenol. Notably, only the 14-3-3-associated Ca V 1.2 population displayed increased cluster size with isoproterenol, revealing a role for 14-3-3 as a nucleation factor that directs Ca V 1.2 super-clustering. 14-3-3 overexpression increased basal Ca V 1.2 cluster size and Ca 2+ currents in ventricular myocytes, with maintained channel responsivity to isoproterenol. In contrast, isoproterenol-stimulated augmentation of sarcolemmal Ca V 1.2 expression and currents in ventricular myocytes were abrogated by 14-3-3 inhibition. These data support a model where 14-3-3 interacts with Ca V 1.2 in a phosphorylation-dependent manner to promote enhanced trafficking/recycling, clustering, and activity during β -adrenergic stimulation. Significance Statement The L-type Ca 2+ channel, Ca V 1.2, plays an essential role in excitation-contraction coupling in the heart and in part regulates the overall strength of contraction during basal and fight- or-flight β -adrenergic signaling conditions. Proteins that modulate the trafficking and/or activity of Ca V 1.2 are interesting both from a physiological and pathological perspective, since alterations in Ca V 1.2 can impact action potential duration and cause arrythmias. A small protein called 14-3-3 regulates other ion channels in the heart and other Ca 2+ channels, but how it may interact with Ca V 1.2 in the heart has never been studied. Examining factors that affect Ca V 1.2 at rest and during β -adrenergic stimulation is crucial for our ability to understand and treat disease and aging conditions where these pathways are altered.
Collapse
|
3
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. PLoS One 2024; 19:e0301063. [PMID: 38995900 PMCID: PMC11244776 DOI: 10.1371/journal.pone.0301063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024] Open
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme calcium/calmodulin-dependent protein kinase II (CaMKII) plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| | - Vernon R. J. Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| |
Collapse
|
4
|
Lucaci AG, Brew WE, Lamanna J, Selberg A, Carnevale V, Moore AR, Kosakovsky Pond SL. The evolution of mammalian Rem2: unraveling the impact of purifying selection and coevolution on protein function, and implications for human disorders. FRONTIERS IN BIOINFORMATICS 2024; 4:1381540. [PMID: 38978817 PMCID: PMC11228553 DOI: 10.3389/fbinf.2024.1381540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/28/2024] [Indexed: 07/10/2024] Open
Abstract
Rad And Gem-Like GTP-Binding Protein 2 (Rem2), a member of the RGK family of Ras-like GTPases, is implicated in Huntington's disease and Long QT Syndrome and is highly expressed in the brain and endocrine cells. We examine the evolutionary history of Rem2 identified in various mammalian species, focusing on the role of purifying selection and coevolution in shaping its sequence and protein structural constraints. Our analysis of Rem2 sequences across 175 mammalian species found evidence for strong purifying selection in 70% of non-invariant codon sites which is characteristic of essential proteins that play critical roles in biological processes and is consistent with Rem2's role in the regulation of neuronal development and function. We inferred epistatic effects in 50 pairs of codon sites in Rem2, some of which are predicted to have deleterious effects on human health. Additionally, we reconstructed the ancestral evolutionary history of mammalian Rem2 using protein structure prediction of extinct and extant sequences which revealed the dynamics of how substitutions that change the gene sequence of Rem2 can impact protein structure in variable regions while maintaining core functional mechanisms. By understanding the selective pressures, protein- and gene - interactions that have shaped the sequence and structure of the Rem2 protein, we gain a stronger understanding of its biological and functional constraints.
Collapse
Affiliation(s)
- Alexander G Lucaci
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
- Weill Cornell Medicine, The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, United States
| | - William E Brew
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Jason Lamanna
- Department of Biology, Temple University, Philadelphia, PA, United States
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA, United States
| | - Avery Selberg
- Department of Biology, Temple University, Philadelphia, PA, United States
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, United States
| | - Vincenzo Carnevale
- Department of Biology, Temple University, Philadelphia, PA, United States
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA, United States
| | - Anna R Moore
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Sergei L Kosakovsky Pond
- Department of Biology, Temple University, Philadelphia, PA, United States
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
5
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584540. [PMID: 38558974 PMCID: PMC10979978 DOI: 10.1101/2024.03.11.584540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme CaMKII plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on runaway synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| | - Vernon R J Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| |
Collapse
|
6
|
Papa A, del Rivero Morfin PJ, Chen BX, Yang L, Katchman AN, Zakharov SI, Liu G, Bohnen MS, Zheng V, Katz M, Subramaniam S, Hirsch JA, Weiss S, Dascal N, Karlin A, Pitt GS, Colecraft HM, Ben-Johny M, Marx SO. A membrane-associated phosphoswitch in Rad controls adrenergic regulation of cardiac calcium channels. J Clin Invest 2024; 134:e176943. [PMID: 38227371 PMCID: PMC10904049 DOI: 10.1172/jci176943] [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: 10/26/2023] [Accepted: 01/11/2024] [Indexed: 01/17/2024] Open
Abstract
The ability to fight or flee from a threat relies on an acute adrenergic surge that augments cardiac output, which is dependent on increased cardiac contractility and heart rate. This cardiac response depends on β-adrenergic-initiated reversal of the small RGK G protein Rad-mediated inhibition of voltage-gated calcium channels (CaV) acting through the Cavβ subunit. Here, we investigate how Rad couples phosphorylation to augmented Ca2+ influx and increased cardiac contraction. We show that reversal required phosphorylation of Ser272 and Ser300 within Rad's polybasic, hydrophobic C-terminal domain (CTD). Phosphorylation of Ser25 and Ser38 in Rad's N-terminal domain (NTD) alone was ineffective. Phosphorylation of Ser272 and Ser300 or the addition of 4 Asp residues to the CTD reduced Rad's association with the negatively charged, cytoplasmic plasmalemmal surface and with CaVβ, even in the absence of CaVα, measured here by FRET. Addition of a posttranslationally prenylated CAAX motif to Rad's C-terminus, which constitutively tethers Rad to the membrane, prevented the physiological and biochemical effects of both phosphorylation and Asp substitution. Thus, dissociation of Rad from the sarcolemma, and consequently from CaVβ, is sufficient for sympathetic upregulation of Ca2+ currents.
Collapse
Affiliation(s)
- Arianne Papa
- Division of Cardiology, Department of Medicine, and
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Pedro J. del Rivero Morfin
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Bi-Xing Chen
- Division of Cardiology, Department of Medicine, and
| | - Lin Yang
- Division of Cardiology, Department of Medicine, and
| | | | | | - Guoxia Liu
- Division of Cardiology, Department of Medicine, and
| | | | - Vivian Zheng
- Division of Cardiology, Department of Medicine, and
| | | | | | - Joel A. Hirsch
- Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Arthur Karlin
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute and Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Henry M. Colecraft
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Pharmacology and Molecular Signaling, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Steven O. Marx
- Division of Cardiology, Department of Medicine, and
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Pharmacology and Molecular Signaling, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
7
|
Gan L, Deng Z, Wei Y, Li H, Zhao L. Decreased expression of GEM in osteoarthritis cartilage regulates chondrogenic differentiation via Wnt/β-catenin signaling. J Orthop Surg Res 2023; 18:751. [PMID: 37794464 PMCID: PMC10548561 DOI: 10.1186/s13018-023-04236-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND GEM (GTP-binding protein overexpressed in skeletal muscle) is one of the atypical small GTPase subfamily members recently identified as a regulator of cell differentiation. Abnormal chondrogenesis coupled with an imbalance in the turnover of cartilaginous matrix formation is highly relevant to the onset and progression of osteoarthritis (OA). However, how GEM regulates chondrogenic differentiation remains unexplored. METHODS Cartilage tissues were obtained from OA patients and graded according to the ORASI and ICRS grading systems. The expression alteration of GEM was detected in the Grade 4 cartilage compared to Grade 0 and verified in OA mimic culture systems. Next, to investigate the specific function of GEM during these processes, we generated a Gem knockdown (Gem-Kd) system by transfecting siRNA targeting Gem into ATDC5 cells. Acan, Col2a1, Sox9, and Wnt target genes of Gem-Kd ATDC5 cells were detected during induction. The transcriptomic sequencing analysis was performed to investigate the mechanism of GEM regulation. Wnt signaling pathways were verified by real-time PCR and immunoblot analysis. Finally, a rescue model generated by treating Gem-KD ATDC5 cells with a Wnt signaling agonist was established to validate the mechanism identified by RNA sequencing analysis. RESULTS A decreased expression of GEM in OA patients' cartilage tissues and OA mimic chondrocytes was observed. While during chondrogenesis differentiation and cartilage matrix formation, the expression of GEM was increased. Gem silencing suppressed chondrogenic differentiation and the expressions of Acan, Col2a1, and Sox9. RNA sequencing analysis revealed that Wnt signaling was downregulated in Gem-Kd cells. Decreased expression of Wnt signaling associated genes and the total β-CATENIN in the nucleus and cytoplasm were observed. The exogenous Wnt activation exhibited reversed effect on Gem loss-of-function cells. CONCLUSION These findings collectively validated that GEM functions as a novel regulator mediating chondrogenic differentiation and cartilage matrix formation through Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Lu Gan
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhonghao Deng
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yiran Wei
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | | | - Liang Zhao
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
8
|
Sun Z, Li Y, Tan X, Liu W, He X, Pan D, Li E, Xu L, Long L. Friend or Foe: Regulation, Downstream Effectors of RRAD in Cancer. Biomolecules 2023; 13:biom13030477. [PMID: 36979412 PMCID: PMC10046484 DOI: 10.3390/biom13030477] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Ras-related associated with diabetes (RRAD), a member of the Ras-related GTPase superfamily, is primarily a cytosolic protein that actives in the plasma membrane. RRAD is highly expressed in type 2 diabetes patients and as a biomarker of congestive heart failure. Mounting evidence showed that RRAD is important for the progression and metastasis of tumor cells, which play opposite roles as an oncogene or tumor suppressor gene depending on cancer and cell type. These findings are of great significance, especially given that relevant molecular mechanisms are being discovered. Being regulated in various pathways, RRAD plays wide spectrum cellular activity including tumor cell division, motility, apoptosis, and energy metabolism by modulating tumor-related gene expression and interacting with multiple downstream effectors. Additionally, RRAD in senescence may contribute to its role in cancer. Despite the twofold characters of RRAD, targeted therapies are becoming a potential therapeutic strategy to combat cancers. This review will discuss the dual identity of RRAD in specific cancer type, provides an overview of the regulation and downstream effectors of RRAD to offer valuable insights for readers, explore the intracellular role of RRAD in cancer, and give a reference for future mechanistic studies.
Collapse
Affiliation(s)
- Zhangyue Sun
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Yongkang Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Xiaolu Tan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Wanyi Liu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Xinglin He
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Deyuan Pan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Liyan Xu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Lin Long
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
- Correspondence: ; Tel.: +86-754-88900460; Fax: +86-754-88900847
| |
Collapse
|
9
|
Del Rivero Morfin PJ, Marx SO, Ben-Johny M. Sympathetic Nervous System Regulation of Cardiac Calcium Channels. Handb Exp Pharmacol 2023. [PMID: 36592229 DOI: 10.1007/164_2022_632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Calcium influx through voltage-gated calcium channels, Cav1.2, in cardiomyocytes initiates excitation-contraction coupling in the heart. The force and rate of cardiac contraction are modulated by the sympathetic nervous system, mediated substantially by changes in intracellular calcium. Norepinephrine released from sympathetic neurons innervating the heart and epinephrine secreted by the adrenal chromaffin cells bind to β-adrenergic receptors on the sarcolemma of cardiomyocytes initiating a signaling cascade that generates cAMP and activates protein kinase A, the targets of which control calcium influx. For decades, the mechanisms by which PKA regulated calcium channels in the heart were not known. Recently, these mechanisms have been elucidated. In this chapter, we will review the history of the field and the studies that led to the identification of the evolutionarily conserved process.
Collapse
Affiliation(s)
- Pedro J Del Rivero Morfin
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA. .,Department of Pharmacology and Molecular Signaling, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
10
|
Hu XQ, Zhang L. Oxidative Regulation of Vascular Ca v1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11122432. [PMID: 36552639 PMCID: PMC9774363 DOI: 10.3390/antiox11122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca2+ (Cav1.2) 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 Cav1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav1.2 and potential roles of ROS-mediated Cav1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia.
Collapse
|
11
|
Rio-Vilariño A, del Puerto-Nevado L, García-Foncillas J, Cebrián A. Ras Family of Small GTPases in CRC: New Perspectives for Overcoming Drug Resistance. Cancers (Basel) 2021; 13:3757. [PMID: 34359657 PMCID: PMC8345156 DOI: 10.3390/cancers13153757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer remains among the cancers with the highest incidence, prevalence, and mortality worldwide. Although the development of targeted therapies against the EGFR and VEGFR membrane receptors has considerably improved survival in these patients, the appearance of resistance means that their success is still limited. Overactivation of several members of the Ras-GTPase family is one of the main actors in both tumour progression and the lack of response to cytotoxic and targeted therapies. This fact has led many resources to be devoted over the last decades to the development of targeted therapies against these proteins. However, they have not been as successful as expected in their move to the clinic so far. In this review, we will analyse the role of these Ras-GTPases in the emergence and development of colorectal cancer and their relationship with resistance to targeted therapies, as well as the status and new advances in the design of targeted therapies against these proteins and their possible clinical implications.
Collapse
Affiliation(s)
| | | | - Jesús García-Foncillas
- Translational Oncology Division, Hospital Universitario Fundación Jimenez Diaz, 28040 Madrid, Spain; (A.R.-V.); (L.d.P.-N.)
| | - Arancha Cebrián
- Translational Oncology Division, Hospital Universitario Fundación Jimenez Diaz, 28040 Madrid, Spain; (A.R.-V.); (L.d.P.-N.)
| |
Collapse
|
12
|
Ahern BM, Sebastian A, Levitan BM, Goh J, Andres DA, Satin J. L-type channel inactivation balances the increased peak calcium current due to absence of Rad in cardiomyocytes. J Gen Physiol 2021; 153:212476. [PMID: 34269819 PMCID: PMC8289690 DOI: 10.1085/jgp.202012854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
The L-type Ca2+ channel (LTCC) provides trigger calcium to initiate cardiac contraction in a graded fashion that is regulated by L-type calcium current (ICa,L) amplitude and kinetics. Inactivation of LTCC is controlled to fine-tune calcium flux and is governed by voltage-dependent inactivation (VDI) and calcium-dependent inactivation (CDI). Rad is a monomeric G protein that regulates ICa,L and has recently been shown to be critical to β-adrenergic receptor (β-AR) modulation of ICa,L. Our previous work showed that cardiomyocyte-specific Rad knockout (cRadKO) resulted in elevated systolic function, underpinned by an increase in peak ICa,L, but without pathological remodeling. Here, we sought to test whether Rad-depleted LTCC contributes to the fight-or-flight response independently of β-AR function, resulting in ICa,L kinetic modifications to homeostatically balance cardiomyocyte function. We recorded whole-cell ICa,L from ventricular cardiomyocytes from inducible cRadKO and control (CTRL) mice. The kinetics of ICa,L stimulated with isoproterenol in CTRL cardiomyocytes were indistinguishable from those of unstimulated cRadKO cardiomyocytes. CDI and VDI are both enhanced in cRadKO cardiomyocytes without differences in action potential duration or QT interval. To confirm that Rad loss modulates LTCC independently of β-AR stimulation, we crossed a β1,β2-AR double-knockout mouse with cRadKO, resulting in a Rad-inducible triple-knockout mouse. Deletion of Rad in cardiomyocytes that do not express β1,β2-AR still yielded modulated ICa,L and elevated basal heart function. Thus, in the absence of Rad, increased Ca2+ influx is homeostatically balanced by accelerated CDI and VDI. Our results indicate that the absence of Rad can modulate the LTCC without contribution of β1,β2-AR signaling and that Rad deletion supersedes β-AR signaling to the LTCC to enhance in vivo heart function.
Collapse
Affiliation(s)
- Brooke M Ahern
- Department of Physiology, University of Kentucky, Lexington, KY
| | | | - Bryana M Levitan
- Department of Physiology, University of Kentucky, Lexington, KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
| | - Jensen Goh
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
13
|
Kärki T, Tojkander S. TRPV Protein Family-From Mechanosensing to Cancer Invasion. Biomolecules 2021; 11:1019. [PMID: 34356643 PMCID: PMC8301805 DOI: 10.3390/biom11071019] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Biophysical cues from the cellular microenvironment are detected by mechanosensitive machineries that translate physical signals into biochemical signaling cascades. At the crossroads of extracellular space and cell interior are located several ion channel families, including TRP family proteins, that are triggered by mechanical stimuli and drive intracellular signaling pathways through spatio-temporally controlled Ca2+-influx. Mechanosensitive Ca2+-channels, therefore, act as critical components in the rapid transmission of physical signals into biologically compatible information to impact crucial processes during development, morphogenesis and regeneration. Given the mechanosensitive nature of many of the TRP family channels, they must also respond to the biophysical changes along the development of several pathophysiological conditions and have also been linked to cancer progression. In this review, we will focus on the TRPV, vanilloid family of TRP proteins, and their connection to cancer progression through their mechanosensitive nature.
Collapse
Affiliation(s)
- Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, 00076 Espoo, Finland;
| | - Sari Tojkander
- Department of Veterinary Biosciences, Section of Pathology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
14
|
Reconstitution of β-adrenergic regulation of Ca V1.2: Rad-dependent and Rad-independent protein kinase A mechanisms. Proc Natl Acad Sci U S A 2021; 118:2100021118. [PMID: 34001616 DOI: 10.1073/pnas.2100021118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
L-type voltage-gated CaV1.2 channels crucially regulate cardiac muscle contraction. Activation of β-adrenergic receptors (β-AR) augments contraction via protein kinase A (PKA)-induced increase of calcium influx through CaV1.2 channels. To date, the full β-AR cascade has never been heterologously reconstituted. A recent study identified Rad, a CaV1.2 inhibitory protein, as essential for PKA regulation of CaV1.2. We corroborated this finding and reconstituted the complete pathway with agonist activation of β1-AR or β2-AR in Xenopus oocytes. We found, and distinguished between, two distinct pathways of PKA modulation of CaV1.2: Rad dependent (∼80% of total) and Rad independent. The reconstituted system reproduces the known features of β-AR regulation in cardiomyocytes and reveals several aspects: the differential regulation of posttranslationally modified CaV1.2 variants and the distinct features of β1-AR versus β2-AR activity. This system allows for the addressing of central unresolved issues in the β-AR-CaV1.2 cascade and will facilitate the development of therapies for catecholamine-induced cardiac pathologies.
Collapse
|
15
|
Levitan BM, Ahern BM, Aloysius A, Brown L, Wen Y, Andres DA, Satin J. Rad-GTPase contributes to heart rate via L-type calcium channel regulation. J Mol Cell Cardiol 2021; 154:60-69. [PMID: 33556393 PMCID: PMC8068610 DOI: 10.1016/j.yjmcc.2021.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 01/04/2021] [Accepted: 01/25/2021] [Indexed: 12/19/2022]
Abstract
Sinoatrial node cardiomyocytes (SANcm) possess automatic, rhythmic electrical activity. SAN rate is influenced by autonomic nervous system input, including sympathetic nerve increases of heart rate (HR) via activation of β-adrenergic receptor signaling cascade (β-AR). L-type calcium channel (LTCC) activity contributes to membrane depolarization and is a central target of β-AR signaling. Recent studies revealed that the small G-protein Rad plays a central role in β-adrenergic receptor directed modulation of LTCC. These studies have identified a conserved mechanism in which β-AR stimulation results in PKA-dependent Rad phosphorylation: depletion of Rad from the LTCC complex, which is proposed to relieve the constitutive inhibition of CaV1.2 imposed by Rad association. Here, using a transgenic mouse model permitting conditional cardiomyocyte selective Rad ablation, we examine the contribution of Rad to the control of SANcm LTCC current (ICa,L) and sinus rhythm. Single cell analysis from a recent published database indicates that Rad is expressed in SANcm, and we show that SANcm ICa,L was significantly increased in dispersed SANcm following Rad silencing compared to those from CTRL hearts. Moreover, cRadKO SANcm ICa,L was not further increased with β-AR agonists. We also evaluated heart rhythm in vivo using radiotelemetered ECG recordings in ambulating mice. In vivo, intrinsic HR is significantly elevated in cRadKO. During the sleep phase cRadKO also show elevated HR, and during the active phase there is no significant difference. Rad-deletion had no significant effect on heart rate variability. These results are consistent with Rad governing LTCC function under relatively low sympathetic drive conditions to contribute to slower HR during the diurnal sleep phase HR. In the absence of Rad, the tonic modulated SANcm ICa,L promotes elevated sinus HR. Future novel therapeutics for bradycardia targeting Rad - LTCC can thus elevate HR while retaining βAR responsiveness.
Collapse
Affiliation(s)
- Bryana M Levitan
- Department of Physiology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America; Gill Heart and Vascular Institute, From the University of Kentucky College of Medicine, Lexington, KY, United States of America
| | - Brooke M Ahern
- Department of Physiology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America
| | - Ajoy Aloysius
- Department of Biology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America
| | - Laura Brown
- Department of Physiology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America
| | - Yuan Wen
- Department of Physiology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America; Center for Muscle Biology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, From the University of Kentucky College of Medicine, Lexington, KY, United States of America
| | - Jonathan Satin
- Department of Physiology, From the University of Kentucky College of Medicine, Lexington, KY, United States of America.
| |
Collapse
|
16
|
Man KNM, Bartels P, Horne MC, Hell JW. Tissue-specific adrenergic regulation of the L-type Ca 2+ channel Ca V1.2. Sci Signal 2020; 13:13/663/eabc6438. [PMID: 33443233 DOI: 10.1126/scisignal.abc6438] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ca2+ influx through the L-type Ca2+ channel Cav1.2 triggers each heartbeat. The fight-or-flight response induces the release of the stress response hormone norepinephrine to stimulate β-adrenergic receptors, cAMP production, and protein kinase A activity to augment Ca2+ influx through Cav1.2 and, consequently, cardiomyocyte contractility. Emerging evidence shows that Cav1.2 is regulated by different mechanisms in cardiomyocytes compared to neurons and vascular smooth muscle cells.
Collapse
Affiliation(s)
- Kwun Nok Mimi Man
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Peter Bartels
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Mary C Horne
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA.
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
17
|
Brandalise F, Ratto D, Leone R, Olivero F, Roda E, Locatelli CA, Grazia Bottone M, Rossi P. Deeper and Deeper on the Role of BK and Kir4.1 Channels in Glioblastoma Invasiveness: A Novel Summative Mechanism? Front Neurosci 2020; 14:595664. [PMID: 33328867 PMCID: PMC7734145 DOI: 10.3389/fnins.2020.595664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
In the last decades, increasing evidence has revealed that a large number of channel protein and ion pumps exhibit impaired expression in cancers. This dysregulation is responsible for high proliferative rates as well as migration and invasiveness, reflected in the recently coined term oncochannelopathies. In glioblastoma (GBM), the most invasive and aggressive primary brain tumor, GBM cells modify their ionic equilibrium in order to change their volume as a necessary step prior to migration. This mechanism involves increased expression of BK channels and downregulation of the normally widespread Kir4.1 channels, as noted in GBM biopsies from patients. Despite a large body of work implicating BK channels in migration in response to an artificial intracellular calcium rise, little is known about how this channel acts in GBM cells at resting membrane potential (RMP), as compared to other channels that are constitutively open, such as Kir4.1. In this review we propose that a residual fraction of functionally active Kir4.1 channels mediates a small, but continuous, efflux of potassium at the more depolarized RMP of GBM cells. In addition, coinciding with transient membrane deformation and the intracellular rise in calcium concentration, brief activity of BK channels can induce massive and rapid cytosolic water loss that reduces cell volume (cell shrinkage), a necessary step for migration within the brain parenchyma.
Collapse
Affiliation(s)
- Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Daniela Ratto
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| | - Roberta Leone
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Federico Olivero
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy.,Pavia Poison Centre, National Toxicology Information Centre, Laboratory of Clinical & Experimental Toxicology, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Alessandro Locatelli
- Pavia Poison Centre, National Toxicology Information Centre, Laboratory of Clinical & Experimental Toxicology, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani," University of Pavia, Pavia, Italy
| |
Collapse
|
18
|
Stiegler AL, Boggon TJ. The pseudoGTPase group of pseudoenzymes. FEBS J 2020; 287:4232-4245. [PMID: 32893973 PMCID: PMC7544640 DOI: 10.1111/febs.15554] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022]
Abstract
Pseudoenzymes are emerging as significant mediators and regulators of signal transduction. These proteins maintain enzyme folds and topologies, but are disrupted in the conserved motifs required for enzymatic activity. Among the pseudoenzymes, the pseudoGTPase group of atypical GTPases has recently expanded and includes the Rnd and RGK groups, RhoH and the RhoBTB proteins, mitochondrial RhoGTPase and centaurin-γ groups, CENP-M, dynein LIC, Entamoeba histolytica RabX3, leucine-rich repeat kinase 2, and the p190RhoGAP proteins. The wide range of cellular functions associated with pseudoGTPases includes cell migration and adhesion, membrane trafficking and cargo transport, mitosis, mitochondrial activity, transcriptional control, and autophagy, placing the group in an expanding portfolio of signaling pathways. In this review, we examine how the pseudoGTPases differ from canonical GTPases and consider their mechanistic and functional roles in signal transduction. We review the amino acid differences between the pseudoGTPases and discuss how these proteins can be classified based on their ability to bind nucleotide and their enzymatic activity. We discuss the molecular and structural consequences of amino acid divergence from canonical GTPases and use comparison with the well-studied pseudokinases to illustrate the classifications. PseudoGTPases are fast becoming recognized as important mechanistic components in a range of cellular roles, and we provide a concise discussion of the currently identified members of this group. ENZYMES: small GTPases; EC number: EC 3.6.5.2.
Collapse
Affiliation(s)
- Amy L. Stiegler
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Titus J. Boggon
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Departments of Molecular Biophysics and Biochemistry, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
19
|
Chinigò G, Fiorio Pla A, Gkika D. TRP Channels and Small GTPases Interplay in the Main Hallmarks of Metastatic Cancer. Front Pharmacol 2020; 11:581455. [PMID: 33132914 PMCID: PMC7550629 DOI: 10.3389/fphar.2020.581455] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Transient Receptor Potential (TRP) cations channels, as key regulators of intracellular calcium homeostasis, play a central role in the essential hallmarks of cancer. Among the multiple pathways in which TRPs may be involved, here we focus our attention on the ones involving small guanosine triphosphatases (GTPases), summarizing the main processes associated with the metastatic cascade, such as migration, invasion and tumor vascularization. In the last decade, several studies have highlighted a bidirectional interplay between TRPs and small GTPases in cancer progression: TRP channels may affect small GTPases activity via both Ca2+-dependent or Ca2+-independent pathways, and, conversely, some small GTPases may affect TRP channels activity through the regulation of their intracellular trafficking to the plasma membrane or acting directly on channel gating. In particular, we will describe the interplay between TRPC1, TRPC5, TRPC6, TRPM4, TRPM7 or TRPV4, and Rho-like GTPases in regulating cell migration, the cooperation of TRPM2 and TRPV2 with Rho GTPases in increasing cell invasiveness and finally, the crosstalk between TRPC1, TRPC6, TRPM8, TRPV4 and both Rho- and Ras-like GTPases in inducing aberrant tumor vascularization.
Collapse
Affiliation(s)
- Giorgia Chinigò
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Laboratoire de Cell Physiology, Université de Lille, Department of Life Sciences, Univ. Lille, Inserm, U1003-PHYCEL, Lille, France
| | - Alessandra Fiorio Pla
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Laboratoire de Cell Physiology, Université de Lille, Department of Life Sciences, Univ. Lille, Inserm, U1003-PHYCEL, Lille, France
| | - Dimitra Gkika
- Laboratoire de Cell Physiology, Université de Lille, Department of Life Sciences, Univ. Lille, Inserm, U1003-PHYCEL, Lille, France.,Univ. Lille, CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020-UMR 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
20
|
Pleiotropic Roles of Calmodulin in the Regulation of KRas and Rac1 GTPases: Functional Diversity in Health and Disease. Int J Mol Sci 2020; 21:ijms21103680. [PMID: 32456244 PMCID: PMC7279331 DOI: 10.3390/ijms21103680] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin is a ubiquitous signalling protein that controls many biological processes due to its capacity to interact and/or regulate a large number of cellular proteins and pathways, mostly in a Ca2+-dependent manner. This complex interactome of calmodulin can have pleiotropic molecular consequences, which over the years has made it often difficult to clearly define the contribution of calmodulin in the signal output of specific pathways and overall biological response. Most relevant for this review, the ability of calmodulin to influence the spatiotemporal signalling of several small GTPases, in particular KRas and Rac1, can modulate fundamental biological outcomes such as proliferation and migration. First, direct interaction of calmodulin with these GTPases can alter their subcellular localization and activation state, induce post-translational modifications as well as their ability to interact with effectors. Second, through interaction with a set of calmodulin binding proteins (CaMBPs), calmodulin can control the capacity of several guanine nucleotide exchange factors (GEFs) to promote the switch of inactive KRas and Rac1 to an active conformation. Moreover, Rac1 is also an effector of KRas and both proteins are interconnected as highlighted by the requirement for Rac1 activation in KRas-driven tumourigenesis. In this review, we attempt to summarize the multiple layers how calmodulin can regulate KRas and Rac1 GTPases in a variety of cellular events, with biological consequences and potential for therapeutic opportunities in disease settings, such as cancer.
Collapse
|
21
|
Autism-associated mutations in the CaVβ2 calcium-channel subunit increase Ba2+-currents and lead to differential modulation by the RGK-protein Gem. Neurobiol Dis 2020; 136:104721. [DOI: 10.1016/j.nbd.2019.104721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/06/2019] [Accepted: 12/26/2019] [Indexed: 12/26/2022] Open
|
22
|
Ras associated with diabetes may play a role in fracture nonunion development in rats. BMC Musculoskelet Disord 2019; 20:602. [PMID: 31830958 PMCID: PMC6909478 DOI: 10.1186/s12891-019-2970-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Rad is the prototypic member of a subfamily of Ras-related small G-proteins and is highly expressed in the skeletal muscle of patients with type II diabetes. Our previous microarray analysis suggested that Rad may mediate fracture nonunion development. Thus, the present study used rat experimental models to investigate and compare the gene and protein expression patterns of both Rad and Rem1, another RGK subfamily member, in nonunions and standard healing fractures. Methods Standard healing fractures and nonunions (produced via periosteal cauterization at the fracture site) were created in the femurs of 3-month-old male Sprague-Dawley rats. At post-fracture days 7, 14, 21, and 28, the fracture callus and fibrous tissue from the standard healing fractures and nonunions, respectively, were harvested and screened (via real-time PCR) for Rad and Rem1 expression. The immunolocalization of both encoded proteins was analyzed at post-fracture days 14 and 21. At the same time points, hematoxylin and eosin staining was performed to identify the detailed tissue structures. Results Results of real-time PCR analysis showed that Rad expression increased significantly in the nonunions, compared to that in the standard healing fractures, at post-fracture days 14, 21, and 28. Conversely, immunohistochemical analysis revealed the immunolocalization of Rad to be similar to that of Rem1 in both fracture types at post-fracture days 14 and 21. Conclusions Rad may mediate nonunion development, and thus, may be a promising therapeutic target to treat these injuries.
Collapse
|
23
|
Morgenstern TJ, Park J, Fan QR, Colecraft HM. A potent voltage-gated calcium channel inhibitor engineered from a nanobody targeted to auxiliary Ca Vβ subunits. eLife 2019; 8:49253. [PMID: 31403402 PMCID: PMC6701945 DOI: 10.7554/elife.49253] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/10/2019] [Indexed: 12/15/2022] Open
Abstract
Inhibiting high-voltage-activated calcium channels (HVACCs; CaV1/CaV2) is therapeutic for myriad cardiovascular and neurological diseases. For particular applications, genetically-encoded HVACC blockers may enable channel inhibition with greater tissue-specificity and versatility than is achievable with small molecules. Here, we engineered a genetically-encoded HVACC inhibitor by first isolating an immunized llama nanobody (nb.F3) that binds auxiliary HVACC CaVβ subunits. Nb.F3 by itself is functionally inert, providing a convenient vehicle to target active moieties to CaVβ-associated channels. Nb.F3 fused to the catalytic HECT domain of Nedd4L (CaV-aβlator), an E3 ubiquitin ligase, ablated currents from diverse HVACCs reconstituted in HEK293 cells, and from endogenous CaV1/CaV2 channels in mammalian cardiomyocytes, dorsal root ganglion neurons, and pancreatic β cells. In cardiomyocytes, CaV-aβlator redistributed CaV1.2 channels from dyads to Rab-7-positive late endosomes. This work introduces CaV-aβlator as a potent genetically-encoded HVACC inhibitor, and describes a general approach that can be broadly adapted to generate versatile modulators for macro-molecular membrane protein complexes.
Collapse
Affiliation(s)
- Travis J Morgenstern
- Department of Pharmacology, Columbia University, Vagelos College of Physicians and Surgeons, New York, United States
| | - Jinseo Park
- Department of Pharmacology, Columbia University, Vagelos College of Physicians and Surgeons, New York, United States
| | - Qing R Fan
- Department of Pharmacology, Columbia University, Vagelos College of Physicians and Surgeons, New York, United States
| | - Henry M Colecraft
- Department of Pharmacology, Columbia University, Vagelos College of Physicians and Surgeons, New York, United States.,Department of Physiology and Cellular Biophysics, Columbia University, Vagelos College of Physicians and Surgeons, New York, United States
| |
Collapse
|
24
|
Wang Z, Feng M, Awe O, Ma Y, Shen M, Xue P, Ahima R, Wolfe A, Segars J, Wu S. Gonadotrope androgen receptor mediates pituitary responsiveness to hormones and androgen-induced subfertility. JCI Insight 2019; 5:127817. [PMID: 31393859 DOI: 10.1172/jci.insight.127817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Many women with hyperandrogenemia suffer from irregular menses and infertility. However, it is unknown whether androgens directly affect reproduction. Since animal models of hyperandrogenemia-induced infertility are associated with obesity, which may impact reproductive function, we have created a lean mouse model of elevated androgen using implantation of low dose dihydrotestosterone (DHT) pellets to separate the effects of elevated androgen from obesity. The hypothalamic-pituitary-gonadal axis controls reproduction. While we have demonstrated that androgen impairs ovarian function, androgen could also disrupt neuroendocrine function at the level of brain and/or pituitary to cause infertility. To understand how elevated androgens might act on pituitary gonadotropes to influence reproductive function, female mice with disruption of the androgen receptor (Ar) gene specifically in pituitary gonadotropes (PitARKO) were produced. DHT treated control mice with intact pituitary Ar (Con-DHT) exhibit disrupted estrous cyclicity and fertility with reduced pituitary responsiveness to GnRH at the level of both calcium signaling and LH secretion. These effects were ameliorated in DHT treated PitARKO mice. Calcium signaling controls GnRH regulation of LH vesicle exotocysis. Our data implicated upregulation of GEM (a voltage-dependent calcium channel inhibitor) in the pituitary as a potential mechanism for androgen's pathological effects. These results demonstrate that gonadotrope AR, as an extra-ovarian regulator, plays an important role in reproductive pathophysiology.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mingxiao Feng
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olubusayo Awe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yaping Ma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Mingjie Shen
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Gynecology and Obstetrics, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Xue
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Molecular and Cellular Physiology, and
| | - James Segars
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Molecular and Cellular Physiology, and.,Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Ahern BM, Levitan BM, Veeranki S, Shah M, Ali N, Sebastian A, Su W, Gong MC, Li J, Stelzer JE, Andres DA, Satin J. Myocardial-restricted ablation of the GTPase RAD results in a pro-adaptive heart response in mice. J Biol Chem 2019; 294:10913-10927. [PMID: 31147441 PMCID: PMC6635439 DOI: 10.1074/jbc.ra119.008782] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/16/2019] [Indexed: 12/25/2022] Open
Abstract
Existing therapies to improve heart function target β-adrenergic receptor (β-AR) signaling and Ca2+ handling and often lead to adverse outcomes. This underscores an unmet need for positive inotropes that improve heart function without any adverse effects. The GTPase Ras associated with diabetes (RAD) regulates L-type Ca2+ channel (LTCC) current (ICa,L). Global RAD-knockout mice (gRAD-/-) have elevated Ca2+ handling and increased cardiac hypertrophy, but RAD is expressed also in noncardiac tissues, suggesting the possibility that pathological remodeling is due also to noncardiac effects. Here, we engineered a myocardial-restricted inducible RAD-knockout mouse (RADΔ/Δ). Using an array of methods and techniques, including single-cell electrophysiological and calcium transient recordings, echocardiography, and radiotelemetry monitoring, we found that RAD deficiency results in a sustained increase of inotropy without structural or functional remodeling of the heart. ICa,L was significantly increased, with RAD loss conferring a β-AR-modulated phenotype on basal ICa,L Cardiomyocytes from RADΔ/Δ hearts exhibited enhanced cytosolic Ca2+ handling, increased contractile function, elevated sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2a) expression, and faster lusitropy. These results argue that myocardial RAD ablation promotes a beneficial elevation in Ca2+ dynamics, which would obviate a need for increased β-AR signaling to improve cardiac function.
Collapse
Affiliation(s)
| | - Bryana M Levitan
- Department of Physiology,; Gill Heart and Vascular Institute, and
| | - Sudhakar Veeranki
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536 and
| | | | - Nemat Ali
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536 and
| | | | | | | | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536 and.
| | | |
Collapse
|
26
|
Xu Q, Wang Y, Zhu J, Zhao Y, Lin Y. Molecular characterization of GTP binding protein overexpressed in skeletal muscle (GEM) and its role in promoting adipogenesis in goat intramuscular preadipocytes. Anim Biotechnol 2018; 31:17-24. [PMID: 30570352 DOI: 10.1080/10495398.2018.1523796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
GTP binding protein overexpressed in skeletal muscle (GEM) is an important gene with many functions, such as regulating the rearrangement of cytoskeleton and the activity of voltage-dependent calcium channel, and GEM was regarded as a candidate gene for obesity. However, little investigation has been carried out to explore whether GEM affected the intramuscular fat (IMF) deposition of goat. To explore the role of GEM gene in goat, this gene was cloned and its tissue and temporal expression profile were detected. Effect of GEM on adipogenesis was examined by losing function of GEM in vitro. Thereafter, several lipid metabolism-related genes were examined, including CCAAT/enhancing-binding protein α (C/EBPα), CCAAT/enhancing-binding protein β (C/EBPβ), lipoprotein lipase (LPL), preadipocyte factor 1 (Pref-1), peroxisome proliferator activated receptor γ (PPARγ) and sterol regulatory element binding protein 1 (SREBP1). We found that the goat GEM gene consisted of 936 bp, which encoded a protein of 311 amino acids. The expression of GEM was higher in spleen, lung and large intestine and it appeared sharp in the interim stage of differentiation. Furthermore, GEM knockdown blocked adipogenesis and the expression of C/EBPα, C/EBPβ, LPL, PPARγ and SREBP1. These results indicated that GEM might promote lipid accumulation and adipogenesis.
Collapse
Affiliation(s)
- Qing Xu
- School of Life Science and Technology, Southwest Minzu University, Chengdu, P.R. China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu UniversityChengdu, Sichuan, P.R. China
| | - Yong Wang
- School of Life Science and Technology, Southwest Minzu University, Chengdu, P.R. China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu UniversityChengdu, Sichuan, P.R. China.,Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Chengdu, Sichuan, P.R. China
| | - Jiangjiang Zhu
- School of Life Science and Technology, Southwest Minzu University, Chengdu, P.R. China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu UniversityChengdu, Sichuan, P.R. China.,Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Chengdu, Sichuan, P.R. China
| | - Yanying Zhao
- School of Life Science and Technology, Southwest Minzu University, Chengdu, P.R. China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu UniversityChengdu, Sichuan, P.R. China
| | - Yaqiu Lin
- School of Life Science and Technology, Southwest Minzu University, Chengdu, P.R. China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu UniversityChengdu, Sichuan, P.R. China
| |
Collapse
|
27
|
Royer L, Herzog JJ, Kenny K, Tzvetkova B, Cochrane JC, Marr MT, Paradis S. The Ras-like GTPase Rem2 is a potent inhibitor of calcium/calmodulin-dependent kinase II activity. J Biol Chem 2018; 293:14798-14811. [PMID: 30072381 DOI: 10.1074/jbc.ra118.003560] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/20/2018] [Indexed: 02/05/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a well-characterized, abundant protein kinase that regulates a diverse set of functions in a tissue-specific manner. For example, in heart muscle, CaMKII regulates Ca2+ homeostasis, whereas in neurons, CaMKII regulates activity-dependent dendritic remodeling and long-term potentiation (LTP), a neurobiological correlate of learning and memory. Previously, we identified the GTPase Rem2 as a critical regulator of dendrite branching and homeostatic plasticity in the vertebrate nervous system. Here, we report that Rem2 directly interacts with CaMKII and potently inhibits the activity of the intact holoenzyme, a previously unknown Rem2 function. Our results suggest that Rem2 inhibition involves interaction with both the CaMKII hub domain and substrate recognition domain. Moreover, we found that Rem2-mediated inhibition of CaMKII regulates dendritic branching in cultured hippocampal neurons. Lastly, we report that substitution of two key amino acid residues in the Rem2 N terminus (Arg-79 and Arg-80) completely abolishes its ability to inhibit CaMKII. We propose that our biochemical findings will enable further studies unraveling the functional significance of Rem2 inhibition of CaMKII in cells.
Collapse
Affiliation(s)
| | | | | | | | - Jesse C Cochrane
- Department of Molecular Biology and Genetics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Michael T Marr
- From the Department of Biology, .,Rosenstiel Basic Medical Sciences Research Center
| | - Suzanne Paradis
- From the Department of Biology, .,Volen Center for Complex Systems, and.,National Center for Behavioral Genomics, Brandeis University, Waltham, Massachusetts 02454 and
| |
Collapse
|
28
|
Moore AR, Richards SE, Kenny K, Royer L, Chan U, Flavahan K, Van Hooser SD, Paradis S. Rem2 stabilizes intrinsic excitability and spontaneous firing in visual circuits. eLife 2018; 7:e33092. [PMID: 29809135 PMCID: PMC6010341 DOI: 10.7554/elife.33092] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
Abstract
Sensory experience plays an important role in shaping neural circuitry by affecting the synaptic connectivity and intrinsic properties of individual neurons. Identifying the molecular players responsible for converting external stimuli into altered neuronal output remains a crucial step in understanding experience-dependent plasticity and circuit function. Here, we investigate the role of the activity-regulated, non-canonical Ras-like GTPase Rem2 in visual circuit plasticity. We demonstrate that Rem2-/- mice fail to exhibit normal ocular dominance plasticity during the critical period. At the cellular level, our data establish a cell-autonomous role for Rem2 in regulating intrinsic excitability of layer 2/3 pyramidal neurons, prior to changes in synaptic function. Consistent with these findings, both in vitro and in vivo recordings reveal increased spontaneous firing rates in the absence of Rem2. Taken together, our data demonstrate that Rem2 is a key molecule that regulates neuronal excitability and circuit function in the context of changing sensory experience.
Collapse
Affiliation(s)
- Anna R Moore
- Department of BiologyBrandeis UniversityWalthamUnited States
| | - Sarah E Richards
- Department of BiologyBrandeis UniversityWalthamUnited States
- Volen Center for Complex SystemsBrandeis UniversityWalthamUnited States
| | - Katelyn Kenny
- National Center for Behavioral GenomicsBrandeis UniversityWalthamUnited States
| | - Leandro Royer
- Department of BiologyBrandeis UniversityWalthamUnited States
| | - Urann Chan
- Department of BiologyBrandeis UniversityWalthamUnited States
| | - Kelly Flavahan
- Department of BiologyBrandeis UniversityWalthamUnited States
| | - Stephen D Van Hooser
- Department of BiologyBrandeis UniversityWalthamUnited States
- Volen Center for Complex SystemsBrandeis UniversityWalthamUnited States
| | - Suzanne Paradis
- Department of BiologyBrandeis UniversityWalthamUnited States
- Volen Center for Complex SystemsBrandeis UniversityWalthamUnited States
- National Center for Behavioral GenomicsBrandeis UniversityWalthamUnited States
| |
Collapse
|
29
|
Filić V, Marinović M, Šoštar M, Weber I. Modulation of small GTPase activity by NME proteins. J Transl Med 2018; 98:589-601. [PMID: 29434248 DOI: 10.1038/s41374-018-0023-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 12/06/2017] [Accepted: 12/29/2017] [Indexed: 12/26/2022] Open
Abstract
NME proteins are reported to influence signal transduction activity of small GTPases from the Ras superfamily by diverse mechanisms in addition to their generic NDP kinase activity, which replenishes the cytoplasmic pool of GTP. Comprehensive evidence shows that NME proteins modulate the activity of Ras GTPases, in particular members of the Rho family, via binding to their major activators GEFs. Direct interaction between several NMEs and Ras GTPases were also indicated in vitro and in vivo. These modes of regulation are mainly independent of the NME's kinase activity. NMEs also modulate the Ras-mediated signal transduction by interfering with the formation of a Ras signaling complex at the plasma membrane. In several examples, NMEs were proposed to perform the role of GAP proteins by promoting hydrolysis of the bound GTP, but this activity still requires additional verification. Early suggestions that NMEs can activate small GTPases by direct phosphorylation of the bound GDP, or by high-rate loading of GTP onto a closely apposed GTPase, were largely dismissed. In this review article, we survey and put into perspective published examples of identified and hypothetical mechanisms of Ras signaling modulation by NME proteins. We also point out involvement of NMEs in the transcriptional regulation of components of Ras GTPases-mediated signal transduction pathways, and reciprocal regulation of NME function by small GTPases, particularly related to NME's binding to membranes.
Collapse
Affiliation(s)
- Vedrana Filić
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia
| | - Maja Marinović
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia
| | - Marko Šoštar
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia
| | - Igor Weber
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia.
| |
Collapse
|
30
|
Withers CN, Brown DM, Byiringiro I, Allen MR, Condon KW, Satin J, Andres DA. Rad GTPase is essential for the regulation of bone density and bone marrow adipose tissue in mice. Bone 2017; 103:270-280. [PMID: 28732776 PMCID: PMC6886723 DOI: 10.1016/j.bone.2017.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/12/2017] [Accepted: 07/16/2017] [Indexed: 01/03/2023]
Abstract
The small GTP-binding protein Rad (RRAD, Ras associated with diabetes) is the founding member of the RGK (Rad, Rem, Rem2, and Gem/Kir) family that regulates cardiac voltage-gated Ca2+ channel function. However, its cellular and physiological functions outside of the heart remain to be elucidated. Here we report that Rad GTPase function is required for normal bone homeostasis in mice, as Rad deletion results in significantly lower bone mass and higher bone marrow adipose tissue (BMAT) levels. Dynamic histomorphometry in vivo and primary calvarial osteoblast assays in vitro demonstrate that bone formation and osteoblast mineralization rates are depressed, while in vitro osteoclast differentiation is increased, in the absence of Rad. Microarray analysis revealed that canonical osteogenic gene expression (Runx2, osterix, etc.) is not altered in Rad-/- calvarial osteoblasts; instead robust up-regulation of matrix Gla protein (MGP, +11-fold), an inhibitor of extracellular matrix mineralization and a protein secreted during adipocyte differentiation, was observed. Strikingly, Rad deficiency also resulted in significantly higher marrow adipose tissue levels in vivo and promoted spontaneous in vitro adipogenesis of primary calvarial osteoblasts. Adipogenic differentiation of wildtype calvarial osteoblasts resulted in the loss of endogenous Rad protein, further supporting a role for Rad in the control of BMAT levels. These findings reveal a novel in vivo function for Rad and establish a role for Rad signaling in the complex physiological control of skeletal homeostasis and bone marrow adiposity.
Collapse
Affiliation(s)
- Catherine N Withers
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, BBSRB, 741 S Limestone Street, Lexington, KY 40536-0509, USA.
| | - Drew M Brown
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Innocent Byiringiro
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Keith W Condon
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Jonathan Satin
- Department of Physiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298, USA.
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, BBSRB, 741 S Limestone Street, Lexington, KY 40536-0509, USA.
| |
Collapse
|
31
|
Two Components of Aversive Memory in Drosophila, Anesthesia-Sensitive and Anesthesia-Resistant Memory, Require Distinct Domains Within the Rgk1 Small GTPase. J Neurosci 2017; 37:5496-5510. [PMID: 28416593 DOI: 10.1523/jneurosci.3648-16.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/12/2017] [Accepted: 04/12/2017] [Indexed: 12/17/2022] Open
Abstract
Multiple components have been identified that exhibit different stabilities for aversive olfactory memory in Drosophila These components have been defined by behavioral and genetic studies and genes specifically required for a specific component have also been identified. Intermediate-term memory generated after single cycle conditioning is divided into anesthesia-sensitive memory (ASM) and anesthesia-resistant memory (ARM), with the latter being more stable. We determined that the ASM and ARM pathways converged on the Rgk1 small GTPase and that the N-terminal domain-deleted Rgk1 was sufficient for ASM formation, whereas the full-length form was required for ARM formation. Rgk1 is specifically accumulated at the synaptic site of the Kenyon cells (KCs), the intrinsic neurons of the mushroom bodies, which play a pivotal role in olfactory memory formation. A higher than normal Rgk1 level enhanced memory retention, which is consistent with the result that Rgk1 suppressed Rac-dependent memory decay; these findings suggest that rgk1 bolsters ASM via the suppression of forgetting. We propose that Rgk1 plays a pivotal role in the regulation of memory stabilization by serving as a molecular node that resides at KC synapses, where the ASM and ARM pathway may interact.SIGNIFICANCE STATEMENT Memory consists of multiple components. Drosophila olfactory memory serves as a fundamental model with which to investigate the mechanisms that underlie memory formation and has provided genetic and molecular means to identify the components of memory, namely short-term, intermediate-term, and long-term memory, depending on how long the memory lasts. Intermediate memory is further divided into anesthesia-sensitive memory (ASM) and anesthesia-resistant memory (ARM), with the latter being more stable. We have identified a small GTPase in Drosophila, Rgk1, which plays a pivotal role in the regulation of olfactory memory stability. Rgk1 is required for both ASM and ARM. Moreover, N-terminal domain-deleted Rgk1 was sufficient for ASM formation, whereas the full-length form was required for ARM formation.
Collapse
|
32
|
Acquired expression of CblQ367P in mice induces dysplastic myelopoiesis mimicking chronic myelomonocytic leukemia. Blood 2017; 129:2148-2160. [PMID: 28209720 DOI: 10.1182/blood-2016-06-724658] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/07/2017] [Indexed: 12/20/2022] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is a hematological malignancy characterized by uncontrolled proliferation of dysplastic myelomonocytes and frequent progression to acute myeloid leukemia (AML). We identified mutations in the Cbl gene, which encodes a negative regulator of cytokine signaling, in a subset of CMML patients. To investigate the contribution of mutant Cbl in CMML pathogenesis, we generated conditional knockin mice for Cbl that express wild-type Cbl in a steady state and inducibly express CblQ367P , a CMML-associated Cbl mutant. CblQ367P mice exhibited sustained proliferation of myelomonocytes, multilineage dysplasia, and splenomegaly, which are the hallmarks of CMML. The phosphatidylinositol 3-kinase (PI3K)-AKT and JAK-STAT pathways were constitutively activated in CblQ367P hematopoietic stem cells, which promoted cell cycle progression and enhanced chemokine-chemokine receptor activity. Gem, a gene encoding a GTPase that is upregulated by CblQ367P , enhanced hematopoietic stem cell activity and induced myeloid cell proliferation. In addition, Evi1, a gene encoding a transcription factor, was found to cooperate with CblQ367P and progress CMML to AML. Furthermore, targeted inhibition for the PI3K-AKT and JAK-STAT pathways efficiently suppressed the proliferative activity of CblQ367P -bearing CMML cells. Our findings provide insights into the molecular mechanisms underlying mutant Cbl-induced CMML and propose a possible molecular targeting therapy for mutant Cbl-carrying CMML patients.
Collapse
|
33
|
Chang DD, Colecraft HM. Rad and Rem are non-canonical G-proteins with respect to the regulatory role of guanine nucleotide binding in Ca(V)1.2 channel regulation. J Physiol 2016; 593:5075-90. [PMID: 26426338 DOI: 10.1113/jp270889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/27/2015] [Indexed: 12/15/2022] Open
Abstract
Rad and Rem are Ras-like G-proteins linked to diverse cardiovascular functions and pathophysiology. Understanding how Rad and Rem are regulated is important for deepened insights into their pathophysiological roles. As in other Ras-like G-proteins, Rad and Rem contain a conserved guanine-nucleotide binding domain (G-domain). Canonically, G-domains are key control modules, functioning as nucleotide-regulated switches of G-protein activity. Whether Rad and Rem G-domains conform to this canonical paradigm is ambiguous. Here, we used multiple functional measurements in HEK293 cells and cardiomyocytes (Ca(V)1.2 currents, Ca(2+) transients, Ca(V)β binding) as biosensors to probe the role of the G-domain in regulation of Rad and Rem function. We utilized Rad(S105N) and Rem(T94N), which are the cognate mutants to Ras(S17N), a dominant-negative variant of Ras that displays decreased nucleotide binding affinity. In HEK293 cells, over-expression of either Rad(S105N) or Rem(T94N) strongly inhibited reconstituted Ca(V)1.2 currents to the same extent as their wild-type (wt) counterparts, contrasting with reports that Rad(S105N) is functionally inert in HEK293 cells. Adenovirus-mediated expression of either wt Rad or Rad(S105N) in cardiomyocytes dramatically blocked L-type calcium current (I(Ca,L)) and inhibited Ca(2+)-induced Ca(2+) release, contradicting reports that Rad(S105N) acts as a dominant negative in heart. By contrast, Rem(T94N) was significantly less effective than wt Rem at inhibiting I(Ca,L) and Ca(2+) transients in cardiomyocytes. FRET analyses in cardiomyocytes revealed that both Rad(S105N) and Rem(T94N) had moderately reduced binding affinity for Ca(V)βs relative to their wt counterparts. The results indicate Rad and Rem are non-canonical G-proteins with respect to the regulatory role of their G-domain in Ca(V)1.2 regulation.
Collapse
Affiliation(s)
- Donald D Chang
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
34
|
Downs AG, Scholles KR, Hollis DM. Localization of rem2 in the central nervous system of the adult rainbow trout (Oncorhynchus mykiss). J Chem Neuroanat 2016; 78:87-95. [PMID: 27600327 DOI: 10.1016/j.jchemneu.2016.09.001] [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: 07/15/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 11/20/2022]
Abstract
Rem2 is member of the RGK (Rem, Rad, and Gem/Kir) subfamily of the Ras superfamily of GTP binding proteins known to influence Ca2+ entry into the cell. In addition, Rem2, which is found at high levels in the vertebrate brain, is also implicated in cell proliferation and synapse formation. Though the specific, regional localization of Rem2 in the adult mammalian central nervous system has been well-described, such information is lacking in other vertebrates. Rem2 is involved in neuronal processes where the capacities between adults of different vertebrate classes vary. Thus, we sought to localize the rem2 gene in the central nervous system of an adult anamniotic vertebrate, the rainbow trout (Oncorhynchus mykiss). In situ hybridization using a digoxigenin (DIG)-labeled RNA probe was used to identify the regional distribution of rem2 expression throughout the trout central nervous system, while real-time polymerase chain reaction (rtPCR) further supported these findings. Based on in situ hybridization, the regional distribution of rem2 occurred within each major subdivision of the brain and included large populations of rem2 expressing cells in the dorsal telencephalon of the cerebrum, the internal cellular layer of the olfactory bulb, and the optic tectum of the midbrain. In contrast, no rem2 expressing cells were resolved within the cerebellum. These results were corroborated by rtPCR, where differential rem2 expression occurred between the major subdivisions assayed with the highest levels being found in the cerebrum, while it was nearly absent in the cerebellum. These data indicate that rem2 gene expression is broadly distributed and likely influences diverse functions in the adult fish central nervous system.
Collapse
|
35
|
Rusconi F, Ceriotti P, Miragoli M, Carullo P, Salvarani N, Rocchetti M, Di Pasquale E, Rossi S, Tessari M, Caprari S, Cazade M, Kunderfranco P, Chemin J, Bang ML, Polticelli F, Zaza A, Faggian G, Condorelli G, Catalucci D. Peptidomimetic Targeting of Cavβ2 Overcomes Dysregulation of the L-Type Calcium Channel Density and Recovers Cardiac Function. Circulation 2016; 134:534-46. [PMID: 27486162 DOI: 10.1161/circulationaha.116.021347] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND L-type calcium channels (LTCCs) play important roles in regulating cardiomyocyte physiology, which is governed by appropriate LTCC trafficking to and density at the cell surface. Factors influencing the expression, half-life, subcellular trafficking, and gating of LTCCs are therefore critically involved in conditions of cardiac physiology and disease. METHODS Yeast 2-hybrid screenings, biochemical and molecular evaluations, protein interaction assays, fluorescence microscopy, structural molecular modeling, and functional studies were used to investigate the molecular mechanisms through which the LTCC Cavβ2 chaperone regulates channel density at the plasma membrane. RESULTS On the basis of our previous results, we found a direct linear correlation between the total amount of the LTCC pore-forming Cavα1.2 and the Akt-dependent phosphorylation status of Cavβ2 both in a mouse model of diabetic cardiac disease and in 6 diabetic and 7 nondiabetic cardiomyopathy patients with aortic stenosis undergoing aortic valve replacement. Mechanistically, we demonstrate that a conformational change in Cavβ2 triggered by Akt phosphorylation increases LTCC density at the cardiac plasma membrane, and thus the inward calcium current, through a complex pathway involving reduction of Cavα1.2 retrograde trafficking and protein degradation through the prevention of dynamin-mediated LTCC endocytosis; promotion of Cavα1.2 anterograde trafficking by blocking Kir/Gem-dependent sequestration of Cavβ2, thus facilitating the chaperoning of Cavα1.2; and promotion of Cavα1.2 transcription by the prevention of Kir/Gem-mediated shuttling of Cavβ2 to the nucleus, where it limits the transcription of Cavα1.2 through recruitment of the heterochromatin protein 1γ epigenetic repressor to the Cacna1c promoter. On the basis of this mechanism, we developed a novel mimetic peptide that, through targeting of Cavβ2, corrects LTCC life-cycle alterations, facilitating the proper function of cardiac cells. Delivery of mimetic peptide into a mouse model of diabetic cardiac disease associated with LTCC abnormalities restored impaired calcium balance and recovered cardiac function. CONCLUSIONS We have uncovered novel mechanisms modulating LTCC trafficking and life cycle and provide proof of concept for the use of Cavβ2 mimetic peptide as a novel therapeutic tool for the improvement of cardiac conditions correlated with alterations in LTCC levels and function.
Collapse
Affiliation(s)
- Francesca Rusconi
- From Humanitas Clinical and Research Center, Rozzano, Milan, Italy (F.R., P. Ceriotti, M.M., P. Carullo, N.S., E.D.P., P.K., M.-L.B., G.C., D.C.); Institute of Genetic and Biomedical Research UOS Milan National Research Council, Milan, Italy (F.R., P. Carullo, N.S., E.D.P., M.-L.B., D.C.); Department of Biotechnologies and Biosciences, University of Milan-Bicocca, Milan, Italy (M.R., A.Z.); Departments of Life Sciences (S.R.) and Clinical and Experimental Medicine (M.M.), University of Parma, Parma, Italy; University Hospital of Verona, Division of Cardiac Surgery, Verona, Italy (M.T., G.F.); Department of Sciences, University of Roma Tre, Rome, Italy (S.C., F.P.); University of Montpellier, CNRS UMR 5203, INSERM, Department of Neuroscience, Institute for Functional Genomics, LabEx Ion Channel Science and Therapeutics, Montpellier, France (M.C., J.C.); and National Institute of Nuclear Physics, Rome Tre Section, Rome, Italy (F.P.)
| | - Paola Ceriotti
- From Humanitas Clinical and Research Center, Rozzano, Milan, Italy (F.R., P. Ceriotti, M.M., P. Carullo, N.S., E.D.P., P.K., M.-L.B., G.C., D.C.); Institute of Genetic and Biomedical Research UOS Milan National Research Council, Milan, Italy (F.R., P. Carullo, N.S., E.D.P., M.-L.B., D.C.); Department of Biotechnologies and Biosciences, University of Milan-Bicocca, Milan, Italy (M.R., A.Z.); Departments of Life Sciences (S.R.) and Clinical and Experimental Medicine (M.M.), University of Parma, Parma, Italy; University Hospital of Verona, Division of Cardiac Surgery, Verona, Italy (M.T., G.F.); Department of Sciences, University of Roma Tre, Rome, Italy (S.C., F.P.); University of Montpellier, CNRS UMR 5203, INSERM, Department of Neuroscience, Institute for Functional Genomics, LabEx Ion Channel Science and Therapeutics, Montpellier, France (M.C., J.C.); and National Institute of Nuclear Physics, Rome Tre Section, Rome, Italy (F.P.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Liput DJ, Lu VB, Davis MI, Puhl HL, Ikeda SR. Rem2, a member of the RGK family of small GTPases, is enriched in nuclei of the basal ganglia. Sci Rep 2016; 6:25137. [PMID: 27118437 PMCID: PMC4846870 DOI: 10.1038/srep25137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/11/2016] [Indexed: 11/09/2022] Open
Abstract
Rem2 is a member of the RGK subfamily of RAS small GTPases. Rem2 inhibits high voltage activated calcium channels, is involved in synaptogenesis, and regulates dendritic morphology. Rem2 is the primary RGK protein expressed in the nervous system, but to date, the precise expression patterns of this protein are unknown. In this study, we characterized Rem2 expression in the mouse nervous system. In the CNS, Rem2 mRNA was detected in all regions examined, but was enriched in the striatum. An antibody specific for Rem2 was validated using a Rem2 knockout mouse model and used to show abundant expression in striatonigral and striatopallidal medium spiny neurons but not in several interneuron populations. In the PNS, Rem2 was abundant in a subpopulation of neurons in the trigeminal and dorsal root ganglia, but was absent in sympathetic neurons of superior cervical ganglia. Under basal conditions, Rem2 was subject to post-translational phosphorylation, likely at multiple residues. Further, Rem2 mRNA and protein expression peaked at postnatal week two, which corresponds to the period of robust neuronal maturation in rodents. This study will be useful for elucidating the functions of Rem2 in basal ganglia physiology.
Collapse
Affiliation(s)
- Daniel J. Liput
- Laboratories of Molecular Physiology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, 20892-9411, USA
| | - Van B. Lu
- Laboratories of Molecular Physiology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, 20892-9411, USA
| | - Margaret I. Davis
- Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, 20892-9411, USA
| | - Henry L. Puhl
- Laboratories of Molecular Physiology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, 20892-9411, USA
| | - Stephen R. Ikeda
- Laboratories of Molecular Physiology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, 20892-9411, USA
| |
Collapse
|
37
|
Puckerin AA, Chang DD, Subramanyam P, Colecraft HM. Similar molecular determinants on Rem mediate two distinct modes of inhibition of Ca V1.2 channels. Channels (Austin) 2016; 10:379-394. [PMID: 27115600 PMCID: PMC4988437 DOI: 10.1080/19336950.2016.1180489] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rad/Rem/Rem2/Gem (RGK) proteins are Ras-like GTPases that potently inhibit all high-voltage-gated calcium (CaV1/CaV2) channels and are, thus, well-positioned to tune diverse physiological processes. Understanding how RGK proteins inhibit CaV channels is important for perspectives on their (patho)physiological roles and could advance their development and use as genetically-encoded CaV channel blockers. We previously reported that Rem can block surface CaV1.2 channels in 2 independent ways that engage distinct components of the channel complex: (1) by binding auxiliary β subunits (β-binding-dependent inhibition, or BBD); and (2) by binding the pore-forming α1C subunit N-terminus (α1C-binding-dependent inhibition, or ABD). By contrast, Gem uses only the BBD mechanism to block CaV1.2. Rem molecular determinants required for BBD CaV1.2 inhibition are the distal C-terminus and the guanine nucleotide binding G-domain which interact with the plasma membrane and CaVβ, respectively. However, Rem determinants for ABD CaV1.2 inhibition are unknown. Here, combining fluorescence resonance energy transfer, electrophysiology, systematic truncations, and Rem/Gem chimeras we found that the same Rem distal C-terminus and G-domain also mediate ABD CaV1.2 inhibition, but with different interaction partners. Rem distal C-terminus interacts with α1C N-terminus to anchor the G-domain which likely interacts with an as-yet-unidentified site. In contrast to some previous studies, neither the C-terminus of Rem nor Gem was sufficient to inhibit CaV1/CaV2 channels. The results reveal that similar molecular determinants on Rem are repurposed to initiate 2 independent mechanisms of CaV1.2 inhibition.
Collapse
Affiliation(s)
- Akil A Puckerin
- a Department of Pharmacology & Molecular Signaling , Columbia University , New York , NY , USA
| | - Donald D Chang
- b Department of Physiology & Cellular Biophysics , Columbia University , New York , NY , USA
| | - Prakash Subramanyam
- b Department of Physiology & Cellular Biophysics , Columbia University , New York , NY , USA
| | - Henry M Colecraft
- a Department of Pharmacology & Molecular Signaling , Columbia University , New York , NY , USA.,b Department of Physiology & Cellular Biophysics , Columbia University , New York , NY , USA
| |
Collapse
|
38
|
Abstract
According to the standard model of G protein-coupled receptor (GPCR) signaling, GPCRs are localized to the cell membrane where they respond to extracellular signals. Stimulation of GPCRs leads to the activation of heterotrimeric G proteins and their intracellular signaling pathways. However, this model fails to accommodate GPCRs, G proteins, and their downstream effectors that are found on the nuclear membrane or in the nucleus. Evidence from isolated nuclei indicates the presence of GPCRs on the nuclear membrane that can activate similar G protein-dependent signaling pathways in the nucleus as at the cell surface. These pathways also include activation of cyclic adenosine monophosphate, calcium and nitric oxide synthase signaling in cardiomyocytes. In addition, a number of distinct heterotrimeric and monomeric G proteins have been found in the nucleus of various cell types. This review will focus on understanding the function of nuclear G proteins with a focus on cardiac signaling where applicable.
Collapse
|
39
|
van Vugt JJFA, Hoedjes KM, van de Geest HC, Schijlen EWGM, Vet LEM, Smid HM. Differentially expressed genes linked to natural variation in long-term memory formation in Cotesia parasitic wasps. Front Behav Neurosci 2015; 9:255. [PMID: 26557061 PMCID: PMC4617343 DOI: 10.3389/fnbeh.2015.00255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022] Open
Abstract
Even though learning and memory are universal traits in the Animal Kingdom, closely related species reveal substantial variation in learning rate and memory dynamics. To determine the genetic background of this natural variation, we studied two congeneric parasitic wasp species, Cotesia glomerata and C. rubecula, which lay their eggs in caterpillars of the large and small cabbage white butterfly. A successful egg laying event serves as an unconditioned stimulus (US) in a classical conditioning paradigm, where plant odors become associated with the encounter of a suitable host caterpillar. Depending on the host species, the number of conditioning trials and the parasitic wasp species, three different types of transcription-dependent long-term memory (LTM) and one type of transcription-independent, anesthesia-resistant memory (ARM) can be distinguished. To identify transcripts underlying these differences in memory formation, we isolated mRNA from parasitic wasp heads at three different time points between induction and consolidation of each of the four memory types, and for each sample three biological replicates, where after strand-specific paired-end 100 bp deep sequencing. Transcriptomes were assembled de novo and differential expression was determined for each memory type and time point after conditioning, compared to unconditioned wasps. Most differentially expressed (DE) genes and antisense transcripts were only DE in one of the LTM types. Among the DE genes that were DE in two or more LTM types, were many protein kinases and phosphatases, small GTPases, receptors and ion channels. Some genes were DE in opposing directions between any of the LTM memory types and ARM, suggesting that ARM in Cotesia requires the transcription of genes inhibiting LTM or vice versa. We discuss our findings in the context of neuronal functioning, including RNA splicing and transport, epigenetic regulation, neurotransmitter/peptide synthesis and antisense transcription. In conclusion, these brain transcriptomes provide candidate genes that may be involved in the observed natural variation in LTM in closely related Cotesia parasitic wasp species.
Collapse
Affiliation(s)
- Joke J F A van Vugt
- Department of Terrestrial Ecology, Netherlands Institute of Ecology (NIOO-KNAW) Wageningen, Netherlands
| | - Katja M Hoedjes
- Laboratory of Entomology, Wageningen University Wageningen, Netherlands
| | | | - Elio W G M Schijlen
- Applied Bioinformatics, Plant Research International Wageningen, Netherlands
| | - Louise E M Vet
- Department of Terrestrial Ecology, Netherlands Institute of Ecology (NIOO-KNAW) Wageningen, Netherlands ; Laboratory of Entomology, Wageningen University Wageningen, Netherlands
| | - Hans M Smid
- Laboratory of Entomology, Wageningen University Wageningen, Netherlands
| |
Collapse
|
40
|
Campiglio M, Flucher BE. The role of auxiliary subunits for the functional diversity of voltage-gated calcium channels. J Cell Physiol 2015; 230:2019-31. [PMID: 25820299 PMCID: PMC4672716 DOI: 10.1002/jcp.24998] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 03/23/2015] [Indexed: 11/18/2022]
Abstract
Voltage-gated calcium channels (VGCCs) represent the sole mechanism to convert membrane depolarization into cellular functions like secretion, contraction, or gene regulation. VGCCs consist of a pore-forming α1 subunit and several auxiliary channel subunits. These subunits come in multiple isoforms and splice-variants giving rise to a stunning molecular diversity of possible subunit combinations. It is generally believed that specific auxiliary subunits differentially regulate the channels and thereby contribute to the great functional diversity of VGCCs. If auxiliary subunits can associate and dissociate from pre-existing channel complexes, this would allow dynamic regulation of channel properties. However, most auxiliary subunits modulate current properties very similarly, and proof that any cellular calcium channel function is indeed modulated by the physiological exchange of auxiliary subunits is still lacking. In this review we summarize available information supporting a differential modulation of calcium channel functions by exchange of auxiliary subunits, as well as experimental evidence in support of alternative functions of the auxiliary subunits. At the heart of the discussion is the concept that, in their native environment, VGCCs function in the context of macromolecular signaling complexes and that the auxiliary subunits help to orchestrate the diverse protein–protein interactions found in these calcium channel signalosomes. Thus, in addition to a putative differential modulation of current properties, differential subcellular targeting properties and differential protein–protein interactions of the auxiliary subunits may explain the need for their vast molecular diversity. J. Cell. Physiol. 999: 00–00, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc. J. Cell. Physiol. 230: 2019–2031, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marta Campiglio
- Division of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard E Flucher
- Division of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Han Y, Wang L, Yao QP, Zhang P, Liu B, Wang GL, Shen BR, Cheng B, Wang Y, Jiang ZL, Qi YX. Nuclear envelope proteins Nesprin2 and LaminA regulate proliferation and apoptosis of vascular endothelial cells in response to shear stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1165-73. [PMID: 25721888 DOI: 10.1016/j.bbamcr.2015.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/19/2015] [Accepted: 02/15/2015] [Indexed: 11/27/2022]
Abstract
The dysfunction of vascular endothelial cells (ECs) influenced by flow shear stress is crucial for vascular remodeling. However, the roles of nuclear envelope (NE) proteins in shear stress-induced EC dysfunction are still unknown. Our results indicated that, compared with normal shear stress (NSS), low shear stress (LowSS) suppressed the expression of two types of NE proteins, Nesprin2 and LaminA, and increased the proliferation and apoptosis of ECs. Targeted small interfering RNA (siRNA) and gene overexpression plasmid transfection revealed that Nesprin2 and LaminA participate in the regulation of EC proliferation and apoptosis. A protein/DNA array was further used to detect the activation of transcription factors in ECs following transfection with target siRNAs and overexpression plasmids. The regulation of AP-2 and TFIID mediated by Nesprin2 and the activation of Stat-1, Stat-3, Stat-5 and Stat-6 by LaminA were verified under shear stress. Furthermore, using Ingenuity Pathway Analysis software and real-time RT-PCR, the effects of Nesprin2 or LaminA on the downstream target genes of AP-2, TFIID, and Stat-1, Stat-3, Stat-5 and Stat-6, respectively, were investigated under LowSS. Our study has revealed that NE proteins are novel mechano-sensitive molecules in ECs. LowSS suppresses the expression of Nesprin2 and LaminA, which may subsequently modulate the activation of important transcription factors and eventually lead to EC dysfunction.
Collapse
Affiliation(s)
- Yue Han
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Wang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qing-Ping Yao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Liu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Liang Wang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Bao-Rong Shen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Binbin Cheng
- Department of Bioengineering, University of CA, San Diego, USA
| | - Yingxiao Wang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China; Department of Bioengineering, University of CA, San Diego, USA
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
42
|
Buraei Z, Lumen E, Kaur S, Yang J. RGK regulation of voltage-gated calcium channels. SCIENCE CHINA-LIFE SCIENCES 2015; 58:28-38. [PMID: 25576452 PMCID: PMC9074095 DOI: 10.1007/s11427-014-4788-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/02/2014] [Indexed: 01/01/2023]
Abstract
Voltage-gated calcium channels (VGCCs) play critical roles in cardiac and skeletal muscle contractions, hormone and neurotransmitter release, as well as slower processes such as cell proliferation, differentiation, migration and death. Mutations in VGCCs lead to numerous cardiac, muscle and neurological disease, and their physiological function is tightly regulated by kinases, phosphatases, G-proteins, calmodulin and many other proteins. Fifteen years ago, RGK proteins were discovered as the most potent endogenous regulators of VGCCs. They are a family of monomeric GTPases (Rad, Rem, Rem2, and Gem/Kir), in the superfamily of Ras GTPases, and they have two known functions: regulation of cytoskeletal dynamics including dendritic arborization and inhibition of VGCCs. Here we review the mechanisms and molecular determinants of RGK-mediated VGCC inhibition, the physiological impact of this inhibition, and recent evidence linking the two known RGK functions.
Collapse
Affiliation(s)
- Zafir Buraei
- Department of Biology, Pace University, New York, NY, 10038, USA,
| | | | | | | |
Collapse
|
43
|
Xu X, Zhang F, Zamponi GW, Horne WA. Solution NMR and calorimetric analysis of Rem2 binding to the Ca2+ channel β4 subunit: a low affinity interaction is required for inhibition of Cav2.1 Ca2+ currents. FASEB J 2015; 29:1794-804. [PMID: 25563298 DOI: 10.1096/fj.14-264499] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 12/20/2022]
Abstract
Rem, Rad, Kir/Gem (RGK) proteins, including Rem2, mediate profound inhibition of high-voltage activated Ca(2+) channels containing intracellular regulatory β subunits. All RGK proteins bind to voltage-gated Ca(2+) channel β subunit (Cavβ) subunits in vitro, but the necessity of the interaction for current inhibition remains controversial. This study applies NMR and calorimetric techniques to map the binding site for Rem2 on human Cavβ4a and measure its binding affinity. Our experiments revealed 2 binding surfaces on the β4 guanylate kinase domain contributing to a 156 ± 18 µM Kd interaction: a hydrophobic pocket lined by 4 critical residues (L173, N261, H262, and V303), mutation of any of which completely disrupted binding, and a nearby surface containing 3 residues (D206, L209, and D258) that when individually mutated decreased affinity. Voltage-gated Ca(2+) channel α1A subunit (Cav2.1) Ca(2+) currents were completely inhibited by Rem2 when co-expressed with wild-type Cavβ4a, but were unaffected by Rem2 when coexpressed with a Cavβ4a site 1 (L173A/V303A) or site 2 (D258A) mutant. These results provide direct evidence for a low-affinity Rem2/Cavβ4 interaction and show definitively that the interaction is required for Cav2.1 inhibition.
Collapse
Affiliation(s)
- Xingfu Xu
- *Department of Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA; and Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Fangxiong Zhang
- *Department of Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA; and Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald W Zamponi
- *Department of Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA; and Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - William A Horne
- *Department of Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA; and Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
44
|
Andrieu G, Quaranta M, Leprince C, Cuvillier O, Hatzoglou A. Gem GTPase acts upstream Gmip/RhoA to regulate cortical actin remodeling and spindle positioning during early mitosis. Carcinogenesis 2014; 35:2503-11. [PMID: 25173885 DOI: 10.1093/carcin/bgu185] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Gem is a small guanosine triphosphate (GTP)-binding protein within the Ras superfamily, involved in the regulation of voltage-gated calcium channel activity and cytoskeleton reorganization. Gem overexpression leads to stress fiber disruption, actin and cell shape remodeling and neurite elongation in interphase cells. In this study, we show that Gem plays a crucial role in the regulation of cortical actin cytoskeleton that undergoes active remodeling during mitosis. Ectopic expression of Gem leads to cortical actin disruption and spindle mispositioning during metaphase. The regulation of spindle positioning by Gem involves its downstream effector Gmip. Knockdown of Gmip rescued Gem-induced spindle phenotype, although both Gem and Gmip accumulated at the cell cortex. In addition, we implicated RhoA GTPase as an important effector of Gem/Gmip signaling. Inactivation of RhoA by overexpressing dominant-negative mutant prevented normal spindle positioning. Introduction of active RhoA rescued the actin and spindle positioning defects caused by Gem or Gmip overexpression. These findings demonstrate a new role of Gem/Gmip/RhoA signaling in cortical actin regulation during early mitotic stages.
Collapse
Affiliation(s)
- Guillaume Andrieu
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération (LBCMCP), CNRS, F-31062 Toulouse, France, CNRS, Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR 5089, 205 route de Narbonne, BP 64182, F-31077 Toulouse, France and Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - Muriel Quaranta
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération (LBCMCP), CNRS, F-31062 Toulouse, France, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - Corinne Leprince
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération (LBCMCP), CNRS, F-31062 Toulouse, France, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR 5089, 205 route de Narbonne, BP 64182, F-31077 Toulouse, France and Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - Anastassia Hatzoglou
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération (LBCMCP), CNRS, F-31062 Toulouse, France, CNRS, Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR 5089, 205 route de Narbonne, BP 64182, F-31077 Toulouse, France and Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| |
Collapse
|
45
|
Puhl HL, Lu VB, Won YJ, Sasson Y, Hirsch JA, Ono F, Ikeda SR. Ancient origins of RGK protein function: modulation of voltage-gated calcium channels preceded the protostome and deuterostome split. PLoS One 2014; 9:e100694. [PMID: 24992013 PMCID: PMC4081519 DOI: 10.1371/journal.pone.0100694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/23/2014] [Indexed: 11/21/2022] Open
Abstract
RGK proteins, Gem, Rad, Rem1, and Rem2, are members of the Ras superfamily of small GTP-binding proteins that interact with Ca2+ channel β subunits to modify voltage-gated Ca2+ channel function. In addition, RGK proteins affect several cellular processes such as cytoskeletal rearrangement, neuronal dendritic complexity, and synapse formation. To probe the phylogenetic origins of RGK protein–Ca2+ channel interactions, we identified potential RGK-like protein homologs in genomes for genetically diverse organisms from both the deuterostome and protostome animal superphyla. RGK-like protein homologs cloned from Danio rerio (zebrafish) and Drosophila melanogaster (fruit flies) expressed in mammalian sympathetic neurons decreased Ca2+ current density as reported for expression of mammalian RGK proteins. Sequence alignments from evolutionarily diverse organisms spanning the protostome/deuterostome divide revealed conservation of residues within the RGK G-domain involved in RGK protein – Cavβ subunit interaction. In addition, the C-terminal eleven residues were highly conserved and constituted a signature sequence unique to RGK proteins but of unknown function. Taken together, these data suggest that RGK proteins, and the ability to modify Ca2+ channel function, arose from an ancestor predating the protostomes split from deuterostomes approximately 550 million years ago.
Collapse
Affiliation(s)
- Henry L. Puhl
- Laboratory of Molecular Physiology, Section on Transmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - Van B. Lu
- Laboratory of Molecular Physiology, Section on Transmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - Yu-Jin Won
- Laboratory of Molecular Physiology, Section on Transmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - Yehezkel Sasson
- Department of Biochemistry & Molecular Biology, Faculty of Life Sciences, Institute for Structural Biology, Tel Aviv University, Ramat Aviv, Israel
| | - Joel A. Hirsch
- Department of Biochemistry & Molecular Biology, Faculty of Life Sciences, Institute for Structural Biology, Tel Aviv University, Ramat Aviv, Israel
| | - Fumihito Ono
- Laboratory of Molecular Physiology, Section on Model Synaptic Systems, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - Stephen R. Ikeda
- Laboratory of Molecular Physiology, Section on Transmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Ghiretti AE, Paradis S. Molecular mechanisms of activity-dependent changes in dendritic morphology: role of RGK proteins. Trends Neurosci 2014; 37:399-407. [PMID: 24910262 PMCID: PMC4113564 DOI: 10.1016/j.tins.2014.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 01/10/2023]
Abstract
The nervous system has the amazing capacity to transform sensory experience from the environment into changes in neuronal activity that, in turn, cause long-lasting alterations in neuronal morphology. Recent findings indicate that, surprisingly, sensory experience concurrently activates molecular signaling pathways that both promote and inhibit dendritic complexity. Historically, a number of positive regulators of activity-dependent dendritic complexity have been described, whereas the list of identified negative regulators of this process is much shorter. In recent years, there has been an emerging appreciation of the importance of the Rad/Rem/Rem2/Gem/Kir (RGK) GTPases as mediators of activity-dependent structural plasticity. In the following review, we discuss the traditional view of RGK proteins, as well as our evolving understanding of the role of these proteins in instructing structural plasticity.
Collapse
Affiliation(s)
- Amy E Ghiretti
- Department of Biology, National Center for Behavioral Genomics, and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, USA
| | - Suzanne Paradis
- Department of Biology, National Center for Behavioral Genomics, and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
47
|
Neely A, Hidalgo P. Structure-function of proteins interacting with the α1 pore-forming subunit of high-voltage-activated calcium channels. Front Physiol 2014; 5:209. [PMID: 24917826 PMCID: PMC4042065 DOI: 10.3389/fphys.2014.00209] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/15/2014] [Indexed: 11/13/2022] Open
Abstract
Openings of high-voltage-activated (HVA) calcium channels lead to a transient increase in calcium concentration that in turn activate a plethora of cellular functions, including muscle contraction, secretion and gene transcription. To coordinate all these responses calcium channels form supramolecular assemblies containing effectors and regulatory proteins that couple calcium influx to the downstream signal cascades and to feedback elements. According to the original biochemical characterization of skeletal muscle Dihydropyridine receptors, HVA calcium channels are multi-subunit protein complexes consisting of a pore-forming subunit (α1) associated with four additional polypeptide chains β, α2, δ, and γ, often referred to as accessory subunits. Twenty-five years after the first purification of a high-voltage calcium channel, the concept of a flexible stoichiometry to expand the repertoire of mechanisms that regulate calcium channel influx has emerged. Several other proteins have been identified that associate directly with the α1-subunit, including calmodulin and multiple members of the small and large GTPase family. Some of these proteins only interact with a subset of α1-subunits and during specific stages of biogenesis. More strikingly, most of the α1-subunit interacting proteins, such as the β-subunit and small GTPases, regulate both gating and trafficking through a variety of mechanisms. Modulation of channel activity covers almost all biophysical properties of the channel. Likewise, regulation of the number of channels in the plasma membrane is performed by altering the release of the α1-subunit from the endoplasmic reticulum, by reducing its degradation or enhancing its recycling back to the cell surface. In this review, we discuss the structural basis, interplay and functional role of selected proteins that interact with the central pore-forming subunit of HVA calcium channels.
Collapse
Affiliation(s)
- Alan Neely
- Centro Interdisciplinario de Neurociencia de Valparaíso and Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| | - Patricia Hidalgo
- Forschungszentrum Jülich, Institute of Complex Systems 4, Zelluläre Biophysik Jülich, Germany
| |
Collapse
|
48
|
Abstract
Small GTPases are key signal transducers from extracellular stimuli to the nucleus that regulate a variety of cellular responses, including changes in gene expression and cell adhesion and migration. Accumulating data have demonstrated that abnormal activation of these small GTPases plays a critical role in the atherosclerosis characterized by vascular abnormalities, especially endothelial dysfunction and inflammation. Here, we discuss the linkage between small GTPases, inflammation, and atherogenesis. First, small GTPases affect gene expression of inflammatory cytokines through proinflammatory signaling pathways, such as nuclear factor-κB, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, interlukin-8, and monocyte chemoattractant protein-1. Then, these molecules regulate the vascular inflammation through cell adhesion and migration. In turn, small GTPases are also regulated by extracellular stimuli, such as L-selectin, thrombin, oxidized phospholipids, and interleukins. Thus, these inflammatory cytokines generate a vicious cycle for small GTPases and inflammatory responses in the atherogenesis.
Collapse
|
49
|
Scamps F, Sangari S, Bowerman M, Rousset M, Bellis M, Cens T, Charnet P. Nerve injury induces a Gem-GTPase-dependent downregulation of P/Q-type Ca2+ channels contributing to neurite plasticity in dorsal root ganglion neurons. Pflugers Arch 2014; 467:351-66. [PMID: 24809506 DOI: 10.1007/s00424-014-1520-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/08/2014] [Accepted: 04/11/2014] [Indexed: 01/27/2023]
Abstract
Small RGK GTPases, Rad, Gem, Rem1, and Rem2, are potent inhibitors of high-voltage-activated (HVA) Ca(2+) channels expressed in heterologous expression systems. However, the role of this regulation has never been clearly demonstrated in the nervous system. Using transcriptional analysis, we show that peripheral nerve injury specifically upregulates Gem in mice dorsal root ganglia. Following nerve injury, protein expression was increased in ganglia and peripheral nerve, mostly under its phosphorylated form. This was confirmed in situ and in vitro in dorsal root ganglia sensory neurons. Knockdown of endogenous Gem, using specific small-interfering RNA (siRNA), increased the HVA Ca(2+) current only in the large-somatic-sized neurons. Combining pharmacological analysis of the HVA Ca(2+) currents together with Gem siRNA-transfection of larger sensory neurons, we demonstrate that only the P/Q-type Ca(2+) channels were enhanced. In vitro analysis of Gem affinity to various CaVβx-CaV2.x complexes and immunocytochemical studies of Gem and CaVβ expression in sensory neurons suggest that the specific inhibition of the P/Q channels relies on both the regionalized upregulation of Gem and the higher sensitivity of the endogenous CaV2.1-CaVβ4 pair in a subset of sensory neurons including the proprioceptors. Finally, pharmacological inhibition of P/Q-type Ca(2+) current reduces neurite branching of regenerating axotomized neurons. Taken together, the present results indicate that a Gem-dependent P/Q-type Ca(2+) current inhibition may contribute to general homeostatic mechanisms following a peripheral nerve injury.
Collapse
Affiliation(s)
- Frédérique Scamps
- Inserm U1051, Institut des Neurosciences, 80 rue Augustin Fliche, 34091, Montpellier, France,
| | | | | | | | | | | | | |
Collapse
|
50
|
Bio-inspired voltage-dependent calcium channel blockers. Nat Commun 2014; 4:2540. [PMID: 24096474 PMCID: PMC4190111 DOI: 10.1038/ncomms3540] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/03/2013] [Indexed: 12/23/2022] Open
Abstract
Ca2+ influx via voltage-dependent CaV1/CaV2 channels couples electrical signals to biological responses in excitable cells. CaV1/CaV2 channel blockers have broad biotechnological and therapeutic applications. Here we report a general method for developing novel genetically-encoded calcium channel blockers inspired by Rem, a small G-protein that constitutively inhibits CaV1/CaV2 channels. We show that diverse cytosolic proteins (CaVβ, 14-3-3, calmodulin, and CaMKII) that bind pore-forming α1-subunits can be converted into calcium channel blockers with tunable selectivity, kinetics, and potency, simply by anchoring them to the plasma membrane. We term this method “channel inactivation induced by membrane-tethering of an associated protein” (ChIMP). ChIMP is potentially extendable to small-molecule drug discovery, as engineering FK506-binding protein into intracellular sites within CaV1.2-α1C permits heterodimerization-initiated channel inhibition with rapamycin. The results reveal a universal method for developing novel calcium channel blockers that may be extended to develop probes for a broad cohort of unrelated ion channels.
Collapse
|