1
|
Farrell S, Dates J, Ramirez N, Hausknecht-Buss H, Kolosov D. Voltage-gated ion channels are expressed in the Malpighian tubules and anal papillae of the yellow fever mosquito (Aedes aegypti), and may regulate ion transport during salt and water imbalance. J Exp Biol 2024; 227:jeb246486. [PMID: 38197515 PMCID: PMC10912814 DOI: 10.1242/jeb.246486] [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: 07/28/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
Vectors of infectious disease include several species of Aedes mosquitoes. The life cycle of Aedes aegypti, the yellow fever mosquito, consists of a terrestrial adult and an aquatic larval life stage. Developing in coastal waters can expose larvae to fluctuating salinity, causing salt and water imbalance, which is addressed by two prime osmoregulatory organs - the Malpighian tubules (MTs) and anal papillae (AP). Voltage-gated ion channels (VGICs) have recently been implicated in the regulation of ion transport in the osmoregulatory epithelia of insects. In the current study, we: (i) generated MT transcriptomes of freshwater-acclimated and brackish water-exposed larvae of Ae. aegypti, (ii) detected expression of several voltage-gated Ca2+, K+, Na+ and non-ion-selective ion channels in the MTs and AP using transcriptomics, PCR and gel electrophoresis, (iii) demonstrated that mRNA abundance of many altered significantly following brackish water exposure, and (iv) immunolocalized CaV1, NALCN, TRP/Painless and KCNH8 in the MTs and AP of larvae using custom-made antibodies. We found CaV1 to be expressed in the apical membrane of MTs of both larvae and adults, and its inhibition to alter membrane potentials of this osmoregulatory epithelium. Our data demonstrate that multiple VGICs are expressed in osmoregulatory epithelia of Ae. aegypti and may play an important role in the autonomous regulation of ion transport.
Collapse
Affiliation(s)
- Serena Farrell
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Jocelyne Dates
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Nancy Ramirez
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Hannah Hausknecht-Buss
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| | - Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096, USA
| |
Collapse
|
2
|
Monteil A, Guérineau NC, Gil-Nagel A, Parra-Diaz P, Lory P, Senatore A. New insights into the physiology and pathophysiology of the atypical sodium leak channel NALCN. Physiol Rev 2024; 104:399-472. [PMID: 37615954 DOI: 10.1152/physrev.00014.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cell excitability and its modulation by hormones and neurotransmitters involve the concerted action of a large repertoire of membrane proteins, especially ion channels. Unique complements of coexpressed ion channels are exquisitely balanced against each other in different excitable cell types, establishing distinct electrical properties that are tailored for diverse physiological contributions, and dysfunction of any component may induce a disease state. A crucial parameter controlling cell excitability is the resting membrane potential (RMP) set by extra- and intracellular concentrations of ions, mainly Na+, K+, and Cl-, and their passive permeation across the cell membrane through leak ion channels. Indeed, dysregulation of RMP causes significant effects on cellular excitability. This review describes the molecular and physiological properties of the Na+ leak channel NALCN, which associates with its accessory subunits UNC-79, UNC-80, and NLF-1/FAM155 to conduct depolarizing background Na+ currents in various excitable cell types, especially neurons. Studies of animal models clearly demonstrate that NALCN contributes to fundamental physiological processes in the nervous system including the control of respiratory rhythm, circadian rhythm, sleep, and locomotor behavior. Furthermore, dysfunction of NALCN and its subunits is associated with severe pathological states in humans. The critical involvement of NALCN in physiology is now well established, but its study has been hampered by the lack of specific drugs that can block or agonize NALCN currents in vitro and in vivo. Molecular tools and animal models are now available to accelerate our understanding of how NALCN contributes to key physiological functions and the development of novel therapies for NALCN channelopathies.
Collapse
Affiliation(s)
- Arnaud Monteil
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Antonio Gil-Nagel
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Paloma Parra-Diaz
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
3
|
Cobb-Lewis DE, Sansalone L, Khaliq ZM. Contributions of the Sodium Leak Channel NALCN to Pacemaking of Medial Ventral Tegmental Area and Substantia Nigra Dopaminergic Neurons. J Neurosci 2023; 43:6841-6853. [PMID: 37640554 PMCID: PMC10573758 DOI: 10.1523/jneurosci.0930-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023] Open
Abstract
We tested the role of the sodium leak channel, NALCN, in pacemaking of dopaminergic neuron (DAN) subpopulations from adult male and female mice. In situ hybridization revealed NALCN RNA in all DANs, with lower abundance in medial ventral tegmental area (VTA) relative to substantia nigra pars compacta (SNc). Despite lower relative abundance of NALCN, we found that acute pharmacological blockade of NALCN in medial VTA DANs slowed pacemaking by 49.08%. We also examined the electrophysiological properties of projection-defined VTA DAN subpopulations identified by retrograde labeling. Inhibition of NALCN reduced pacemaking in DANs projecting to medial nucleus accumbens (NAc) and others projecting to lateral NAc by 70.74% and 31.98%, respectively, suggesting that NALCN is a primary driver of pacemaking in VTA DANs. In SNc DANs, potentiating NALCN by lowering extracellular calcium concentration speeded pacemaking in wildtype but not NALCN conditional knockout mice, demonstrating functional presence of NALCN. In contrast to VTA DANs, however, pacemaking in SNc DANs was unaffected by inhibition of NALCN. Instead, we found that inhibition of NALCN increased the gain of frequency-current plots at firing frequencies slower than spontaneous firing. Similarly, inhibition of the hyperpolarization-activated cyclic nucleotide-gated (HCN) conductance increased gain but had little effect on pacemaking. Interestingly, simultaneous inhibition of NALCN and HCN resulted in significant reduction in pacemaker rate. Thus, we found NALCN makes substantial contributions to driving pacemaking in VTA DAN subpopulations. In SNc DANs, NALCN is not critical for pacemaking but inhibition of NALCN makes cells more sensitive to hyperpolarizing stimuli.SIGNIFICANCE STATEMENT Pacemaking in midbrain dopaminergic neurons (DAN) relies on multiple subthreshold conductances, including a sodium leak. Whether the sodium leak channel, NALCN, contributes to pacemaking in DANs located in the VTA and the SNc has not yet been determined. Using electrophysiology and pharmacology, we show that NALCN plays a prominent role in driving pacemaking in projection-defined VTA DAN subpopulations. By contrast, pacemaking in SNc neurons does not rely on NALCN. Instead, the presence of NALCN regulates the excitability of SNc DANs by reducing the gain of the neuron's response to inhibitory stimuli. Together, these findings will inform future efforts to obtain DAN subpopulation-specific treatments for use in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dana E Cobb-Lewis
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
- Institute for Neuroscience, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Lorenzo Sansalone
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| | - Zayd M Khaliq
- Cellular Neurophysiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
4
|
Li P, Kuo N, Patel R, Omary MB. Hypoosmosis alters hepatocyte mitochondrial morphology and induces selective release of carbamoyl phosphate synthetase 1. Am J Physiol Gastrointest Liver Physiol 2023; 325:G334-G346. [PMID: 37489865 PMCID: PMC10642991 DOI: 10.1152/ajpgi.00018.2023] [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: 02/07/2023] [Revised: 06/20/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Carbamoyl phosphate synthetase 1 (CPS1) is the most abundant hepatocyte mitochondrial matrix protein. Hypoosmotic stress increases CPS1 release in isolated mouse hepatocytes without cell death. We hypothesized that increased CPS1 release during hypoosmosis is selective and associates with altered mitochondrial morphology. Both ex vivo and in vivo models were assessed. Mouse hepatocytes and livers were challenged with isotonic or hypoosmotic (35 mosM) buffer. Mice were injected intraperitoneally with water (10% body weight) with or without an antidiuretic. Mitochondrial and cytosolic fractions were isolated using differential centrifugation, then analyzed by immunoblotting to assess subcellular redistribution of four mitochondrial proteins: CPS1, ornithine transcarbamylase (OTC), pyrroline-5-carboxylate reductase 1 (PYCR1), and cytochrome c. Mitochondrial morphology alterations were examined using electron microscopy. Hypoosmotic treatment of whole livers or hepatocytes led to preferential or increased mitochondrial release, respectively, of CPS1 as compared with two mitochondrial matrix proteins (OTC/PYCR1) and with the intermembrane space protein, cytochrome c. Mitochondrial apoptosis-induced channel opening using staurosporine in hepatocytes led to preferential CPS1 and cytochrome c release. The CPS1-selective changes were accompanied by dramatic alterations in ultrastructural mitochondrial morphology. In mice, hypoosmosis/hyponatremia led to increased liver vascular congestion and increased CPS1 in bile but not blood, coupled with mitochondrial structural alterations. In contrast, isotonic increase of intravascular volume led to a decrease in mitochondrial size with limited change in bile CPS1 compared with hypoosmotic conditions and absence of the hypoosmosis-associated histological alterations. Taken together, hepatocyte CPS1 is selectively released in response to hypoosmosis/hyponatremia and provides a unique biomarker of mitochondrial injury.NEW & NOTEWORTHY Exposure of isolated mouse livers, primary cultured hepatocytes, or mice to hypoosmosis/hyponatremia conditions induces significant mitochondrial shape alterations accompanied by preferential release of the mitochondrial matrix protein CPS1, a urea cycle enzyme. In contrast, the intermembrane space protein, cytochrome c, and two other matrix proteins, including the urea cycle enzyme ornithine transcarbamylase, remain preferentially retained in mitochondria. Therefore, hepatocyte CPS1 manifests unique mitochondrial stress response compartmentalization and is a sensitive sensor of mitochondrial hypoosmotic/hyponatremic injury.
Collapse
Affiliation(s)
- Pei Li
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States
| | - Ning Kuo
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States
| | - Rajesh Patel
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States
- Department of Pathology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, United States
| | - M Bishr Omary
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, United States
| |
Collapse
|
5
|
James AD, Leslie TK, Kaggie JD, Wiggins L, Patten L, Murphy O'Duinn J, Langer S, Labarthe MC, Riemer F, Baxter G, McLean MA, Gilbert FJ, Kennerley AJ, Brackenbury WJ. Sodium accumulation in breast cancer predicts malignancy and treatment response. Br J Cancer 2022; 127:337-349. [PMID: 35462561 PMCID: PMC9296657 DOI: 10.1038/s41416-022-01802-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer remains a leading cause of death in women and novel imaging biomarkers are urgently required. Here, we demonstrate the diagnostic and treatment-monitoring potential of non-invasive sodium (23Na) MRI in preclinical models of breast cancer. METHODS Female Rag2-/- Il2rg-/- and Balb/c mice bearing orthotopic breast tumours (MDA-MB-231, EMT6 and 4T1) underwent MRI as part of a randomised, controlled, interventional study. Tumour biology was probed using ex vivo fluorescence microscopy and electrophysiology. RESULTS 23Na MRI revealed elevated sodium concentration ([Na+]) in tumours vs non-tumour regions. Complementary proton-based diffusion-weighted imaging (DWI) linked elevated tumour [Na+] to increased cellularity. Combining 23Na MRI and DWI measurements enabled superior classification accuracy of tumour vs non-tumour regions compared with either parameter alone. Ex vivo assessment of isolated tumour slices confirmed elevated intracellular [Na+] ([Na+]i); extracellular [Na+] ([Na+]e) remained unchanged. Treatment with specific inward Na+ conductance inhibitors (cariporide, eslicarbazepine acetate) did not affect tumour [Na+]. Nonetheless, effective treatment with docetaxel reduced tumour [Na+], whereas DWI measures were unchanged. CONCLUSIONS Orthotopic breast cancer models exhibit elevated tumour [Na+] that is driven by aberrantly elevated [Na+]i. Moreover, 23Na MRI enhances the diagnostic capability of DWI and represents a novel, non-invasive biomarker of treatment response with superior sensitivity compared to DWI alone.
Collapse
Affiliation(s)
- Andrew D James
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | | | - Joshua D Kaggie
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | | | - Lewis Patten
- Department of Mathematics, University of York, York, UK
| | | | - Swen Langer
- Bioscience Technology Facility, Department of Biology, University of York, York, UK
| | | | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre, Haukeland University Hospital Bergen, Bergen, Norway
| | - Gabrielle Baxter
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Mary A McLean
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Fiona J Gilbert
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Aneurin J Kennerley
- York Biomedical Research Institute, University of York, York, UK
- Department of Chemistry, University of York, York, UK
| | - William J Brackenbury
- Department of Biology, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
6
|
Douma LG, Costello HM, Crislip GR, Cheng KY, Lynch IJ, Juffre A, Barral D, Masten S, Roig E, Beguiristain K, Li W, Bratanatawira P, Wingo CS, Gumz ML. Kidney-specific KO of the circadian clock protein PER1 alters renal Na + handling, aldosterone levels, and kidney/adrenal gene expression. Am J Physiol Renal Physiol 2022; 322:F449-F459. [PMID: 35129370 PMCID: PMC9169971 DOI: 10.1152/ajprenal.00385.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
PERIOD 1 (PER1) is a circadian clock transcription factor that is regulated by aldosterone, a hormone that increases blood volume and Na+ retention to increase blood pressure. Male global Per1 knockout (KO) mice develop reduced night/day differences in Na+ excretion in response to a high-salt diet plus desoxycorticosterone pivalate treatment (HS + DOCP), a model of salt-sensitive hypertension. In addition, global Per1 KO mice exhibit higher aldosterone levels on a normal-salt diet. To determine the role of Per1 in the kidney, male kidney-specific Per1 KO (KS-Per1 KO) mice were generated using Ksp-cadherin Cre recombinase to remove exons 2-8 of Per1 in the distal nephron and collecting duct. Male KS-Per1 KO mice have increased Na+ retention but have normal diurnal differences in Na+ excretion in response to HS + DOCP. The increased Na+ retention is associated with altered expression of glucocorticoid and mineralocorticoid receptors, increased serum aldosterone, and increased medullary endothelin-1 compared with control mice. Adrenal gland gene expression analysis revealed that circadian clock and aldosterone synthesis genes have altered expression in KS-Per1 KO mice compared with control mice. These results emphasize the importance of the circadian clock not only in maintaining rhythms of physiological functions but also for adaptability in response to environmental cues, such as HS + DOCP, to maintain overall homeostasis. Given the prevalence of salt-sensitive hypertension in the general population, these findings have important implications for our understanding of how circadian clock proteins regulate homeostasis.NEW & NOTEWORTHY For the first time, we show that knockout of the circadian clock transcription factor PERIOD 1 using kidney-specific cadherin Cre results in increased renal Na+ reabsorption, increased aldosterone levels, and changes in gene expression in both the kidney and adrenal gland. Diurnal changes in renal Na+ excretion were not observed, demonstrating that the clock protein PER1 in the kidney is important for maintaining homeostasis and that this effect may be independent of time of day.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Kit-Yan Cheng
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - I Jeanette Lynch
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - Dominique Barral
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Sarah Masten
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Emilio Roig
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Kevin Beguiristain
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Wendy Li
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Phillip Bratanatawira
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Charles S Wingo
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcolm Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|
7
|
Valle AC, Yeh C, Huang Y. Near Infrared-Activatable Platinum-Decorated Gold Nanostars for Synergistic Photothermal/Ferroptotic Therapy in Combating Cancer Drug Resistance. Adv Healthc Mater 2020; 9:e2000864. [PMID: 32945131 DOI: 10.1002/adhm.202000864] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/15/2020] [Indexed: 12/15/2022]
Abstract
Ferroptotic cell death results from glutathione peroxidase 4 (GPX4) inactivation and/or glutathione (GSH) depletion. Elevated GSH levels are often found in multidrug-resistant (MDR) tumor cells, reducing their sensitivity to chemotherapeutic drugs and the efficacy of treatment. MDR cells also acquire a dependency on GPX4, reducing their oxidative stress and promoting their survival. Therefore, the depletion of GSH and inactivation of GPX4 has the potential to be a superior treatment strategy for MDR tumors. Platinum-decorated gold nanostars (Pt-AuNS) are presented as a novel metal nanoprodrug for ferroptotic therapy against MDR tumors. Under dark conditions, the synthesized Pt-AuNS exhibit negligible levels of toxicity. Upon exposure of the Pt-AuNS to near-infrared (NIR) light, active metallic (Pt and Au) species are released, subsequently inducing cytotoxicity. The mechanism of action is attributed to GSH depletion and GPX4 inactivation, accumulating lipid hydroperoxides, which in turn leads to ferroptosis. In in vivo xenograft, the MDR cancer model confirmed the NIR light-activation of Pt-AuNS prodrugs, resulting in efficient ferroptotic therapeutic action against MDR tumors without long-term side effects. The findings lay the groundwork for using Pt-AuNS prodrugs responsive to NIR light as ferroptosis-inducing agents in chemo-resistant cancer cells and demonstrate their potential for use in future clinical applications.
Collapse
Affiliation(s)
- Andrea C. Valle
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Chih‐Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Yu‐Fen Huang
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
- Institute of Analytical and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| |
Collapse
|
8
|
Hahn S, Kim SW, Um KB, Kim HJ, Park MK. N-benzhydryl quinuclidine compounds are a potent and Src kinase-independent inhibitor of NALCN channels. Br J Pharmacol 2020; 177:3795-3810. [PMID: 32436268 DOI: 10.1111/bph.15104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 05/01/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE NALCN is a Na+ leak, GPCR-activated channel that regulates the resting membrane potential and neuronal excitability. Despite numerous possible roles for NALCN in both normal physiology and disease processes, lack of specific blockers hampers further investigation. EXPERIMENTAL APPROACH The effect of N-benzhydryl quinuclidine compounds on NALCN channels was demonstrated using whole-cell patch-clamp recordings in HEK293T cells overexpressing NALCN and acutely isolated nigral dopaminergic neurons that express NALCN endogenously. Src kinase activity was measured using a Src kinase assay kit, and voltage and current-clamp recordings from nigral dopaminergic neurons were used to measure NALCN currents and membrane potentials. KEY RESULTS N-benzhydryl quinuclidine compounds inhibited NALCN channels without affecting TRPC channels, another important route for Na+ leak. In HEK293T cells overexpressing NALCN, N-benzhydryl quinuclidine compounds potently suppressed muscarinic M3 receptor-activated NALCN currents. Structure-function relationship studies suggest that the quinuclidine ring with a benzhydryl group imparts the ability to inhibit NALCN currents regardless of Src family kinases. Moreover, N-benzhydryl quinuclidine compounds inhibited not only GPCR-activated NALCN currents but also background Na+ leak currents and hyperpolarized the membrane potential in native midbrain dopaminergic neurons that express NALCN endogenously. CONCLUSION AND IMPLICATIONS These findings suggest that N-benzhydryl quinuclidine compounds have a pharmacological potential to directly inhibit NALCN channels and could be a useful tool to investigate functions of NALCN channels.
Collapse
Affiliation(s)
- Suyun Hahn
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - So Woon Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ki Bum Um
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun Jin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Myoung Kyu Park
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
9
|
Djamgoz MBA. Hyponatremia and Cancer Progression: Possible Association with Sodium-Transporting Proteins. Bioelectricity 2020; 2:14-20. [PMID: 34471833 DOI: 10.1089/bioe.2019.0035] [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: 12/17/2022] Open
Abstract
Hyponatremia, the phenomenon of serum sodium level falling below 135 mmol/L, is seen frequently in cancer patients and has been correlated with poor prognosis. Hyponatremia has classically been attributed to the "syndrome of inappropriate antidiuretic hormone secretion," leading to prolonged fluid retention. However, this is unlikely to be the only mechanism. In this study, we advance the hypothesis that upregulation of various sodium-transporting proteins during the cancer process makes a significant contribution to the pathophysiology of cancer-associated hyponatremia. Such sodium-transporting proteins include voltage-gated sodium channels, especially its hypoxia-promoted persistent current, epithelial sodium channels, and transient receptor potential channels. Thus, hyponatremia follows cancer, whereby drop in blood serum level occurs as a result of uptake of sodium from extracellular fluid by cancer cells. Indeed, the sodium content of cancer cells/tissues is higher than normal. In turn, the rise in the intracellular sodium concentration brings about a range of cellular effects, including extracellular acidification that promotes invasiveness and thus leads to poor prognosis. This perspective offers novel therapies for cancer and the associated hyponatremia.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom.,Biotechnology Research Centre, Cyprus International University, Lefkosa, North Cyprus
| |
Collapse
|
10
|
Douma LG, Solocinski K, Holzworth MR, Crislip GR, Masten SH, Miller AH, Cheng KY, Lynch IJ, Cain BD, Wingo CS, Gumz ML. Female C57BL/6J mice lacking the circadian clock protein PER1 are protected from nondipping hypertension. Am J Physiol Regul Integr Comp Physiol 2018; 316:R50-R58. [PMID: 30427705 DOI: 10.1152/ajpregu.00381.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian clock is integral to the maintenance of daily rhythms of many physiological outputs, including blood pressure. Our laboratory has previously demonstrated the importance of the clock protein period 1 (PER1) in blood pressure regulation in male mice. Briefly, a high-salt diet (HS; 4% NaCl) plus injection with the long-acting mineralocorticoid deoxycorticosterone pivalate (DOCP) resulted in nondipping hypertension [<10% difference between night and day blood pressure (BP) in Per1-knockout (KO) mice but not in wild-type (WT) mice]. To date, there have been no studies that have examined the effect of a core circadian gene KO on BP rhythms in female mice. The goal of the present study was to determine whether female Per1-KO mice develop nondipping hypertension in response to HS/DOCP treatment. For the first time, we demonstrate that loss of the circadian clock protein PER1 in female mice does not significantly change mean arterial pressure (MAP) or the BP rhythm relative to female C57BL/6 WT control mice. Both WT and Per1-KO female mice experienced a significant increase in MAP in response to HS/DOCP. Importantly, however, both genotypes maintained a >10% dip in BP on HS/DOCP. This effect is distinct from the nondipping hypertension seen in male Per1-KO mice, demonstrating that the female sex appears to be protective against PER1-mediated nondipping hypertension in response to HS/DOCP. Together, these data suggest that PER1 acts in a sex-dependent manner in the regulation of cardiovascular rhythms.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Kristen Solocinski
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | | | - G Ryan Crislip
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Sarah H Masten
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Amber H Miller
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Kit-Yan Cheng
- Department of Medicine, University of Florida , Gainesville, Florida
| | - I Jeanette Lynch
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida
| | - Charles S Wingo
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida.,Research, North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, University of Florida , Gainesville, Florida.,Department of Biochemistry and Molecular Biology, University of Florida , Gainesville, Florida.,Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| |
Collapse
|
11
|
Oh J, Magnuson A, Benoist C, Pittet MJ, Weissleder R. Age-related tumor growth in mice is related to integrin α 4 in CD8+ T cells. JCI Insight 2018; 3:122961. [PMID: 30385729 DOI: 10.1172/jci.insight.122961] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/27/2018] [Indexed: 11/17/2022] Open
Abstract
Cancer incidence increases with age, but paradoxically, cancers have been found to grow more quickly in young mice compared with aged ones. The cause of differential tumor growth has been debated and, over time, attributed to faster tumor cell proliferation, decreased tumor cell apoptosis, and/or increased angiogenesis in young animals. Despite major advances in our understanding of tumor immunity over the past 2 decades, little attention has been paid to comparing immune cell populations in young and aged mice. Using mouse colon adenocarcinoma model MC38 implanted in young and mature mice, we show that age substantially influences the number of tumor-infiltrating cytotoxic CD8+ T cells, which control cancer progression. The different tumor growth pace in young and mature mice was abrogated in RAG1null mice, which lack mature T and B lymphocytes, and upon selective depletion of endogenous CD8+ cells. Transcriptome analysis further indicated that young mice have decreased levels of the Itga4 gene (CD49d, VLA-4) in tumor-infiltrating lymphocytes when compared with mature mice. Hypothesizing that VLA-4 can have a tumor-protective effect, we depleted the protein, which resulted in accelerated tumor growth in mature mice. These observations may explain the paradoxical growth rates observed in murine cancers, point to the central role of VLA-4 in controlling tumor growth, and open new venues to therapeutic manipulation.
Collapse
Affiliation(s)
- Juhyun Oh
- Center for Systems Biology, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA
| | - Angela Magnuson
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Egan JM, Peterson CA, Fry WM. Lack of current observed in HEK293 cells expressing NALCN channels. BIOCHIMIE OPEN 2018; 6:24-28. [PMID: 29892559 PMCID: PMC5991895 DOI: 10.1016/j.biopen.2018.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 01/31/2018] [Indexed: 11/30/2022]
Abstract
The sodium leak channel NALCN is poorly understood, but is reported as a Na+-permeable, nonselective cation leak channel which regulates resting membrane potential and electrical excitability. Previous work has indicated that NALCN currents can be stimulated by activation of several G protein coupled receptors, including the M3 muscarinic receptor. We undertook a study using voltage clamp electrophysiology to investigate NALCN currents. We compared currents elicited from untransfected control HEK239 cells in response to M3R agonists muscarine or Oxotremorine M to currents elicited from cells transfected with M3R only or the M3R plus NALCN and cDNA encoding accessory proteins UNC-80 and Src. Currents with similar properties were observed in all three groups of cells in response to muscarine agonists, in similar proportions of cells tested, from all three groups of cells. Our findings do not support previous electrophysiological studies suggesting that heterologously expressed NALCN functions as a Na+ leak channel in HEK293 cells. More research will be required to determine the molecular requirements for successful expression of the NALCN channel. NALCN (sodium leak channel, non-selective) is a poorly understood ion channel. Several reports indicate that NALCN current can be recorded from transfected cells. Conflicting reports indicate NALCN currents are simply leaky patch clamp seals. We were unable to record currents attributable to NALCN in transfected HEK293 cells. Our experiments suggest that NALCN does not form channels in HEK293 cells.
Collapse
Affiliation(s)
- Jennifer M Egan
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Colleen A Peterson
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - W Mark Fry
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| |
Collapse
|
13
|
Ma X, Pham VT, Mori H, MacDougald OA, Shah YM, Bodary PF. Iron elevation and adipose tissue remodeling in the epididymal depot of a mouse model of polygenic obesity. PLoS One 2017. [PMID: 28651003 PMCID: PMC5484604 DOI: 10.1371/journal.pone.0179889] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Iron dysregulation is a potential contributor to the pathology of obesity-related metabolic complications. KK/HIJ (KK) mice, a polygenic obese mouse model, have elevated serum iron levels. A subset of KK male mice display a bronzing of epididymal adipose tissue (eAT) associated with >100-fold (p<0.001) higher iron concentration. Methods To further phenotype and characterize the adipose tissue iron overload, 27 male KK mice were evaluated. 14 had bronzing eAT and 13 had normal appearing eAT. Fasting serum and tissues were collected for iron content, qPCR, histology and western blot. Results High iron levels were confirmed in bronzing eAT (High Iron group, HI) versus normal iron level (NI) in normal appearing eAT. Surprisingly, iron levels in subcutaneous and brown adipose depots were not different between the groups (p>0.05). The eAT histology revealed iron retention, macrophage clustering, tissue fibrosis, cell death as well as accumulation of HIF-2α in the high iron eAT. qPCR showed significantly decreased Lep (leptin) and AdipoQ (adiponectin), whereas Tnfα (tumor necrosis factor α), and Slc40a1 (ferroportin) were up-regulated in HI (p<0.05). Elevated HIF-2α, oxidative stress and local insulin signaling loss was also observed. Significance Our data suggest that deposition of iron in adipose tissue is limited to the epididymal depot in male KK mice. A robust adipose tissue remodeling is concomitant with the high iron concentration, which causes local adipose tissue insulin resistance.
Collapse
Affiliation(s)
- Xiaoya Ma
- School of Kinesiology, University of Michigan, 1402 Washington Hts., Ann Arbor, MI, United States of America
- Department of Molecular & Integrative Physiology, Ann Arbor, MI, United States of America
| | - Vinh T. Pham
- School of Kinesiology, University of Michigan, 1402 Washington Hts., Ann Arbor, MI, United States of America
| | - Hiroyuki Mori
- Department of Molecular & Integrative Physiology, Ann Arbor, MI, United States of America
| | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology, Ann Arbor, MI, United States of America
- Internal Medicine, University of Michigan Medical School, Ann Arbor MI, United States of America
| | - Yatrik M. Shah
- Department of Molecular & Integrative Physiology, Ann Arbor, MI, United States of America
- Internal Medicine, University of Michigan Medical School, Ann Arbor MI, United States of America
| | - Peter F. Bodary
- School of Kinesiology, University of Michigan, 1402 Washington Hts., Ann Arbor, MI, United States of America
- * E-mail:
| |
Collapse
|
14
|
Ackert-Bicknell CL, Anderson LC, Sheehan S, Hill WG, Chang B, Churchill GA, Chesler EJ, Korstanje R, Peters LL. Aging Research Using Mouse Models. ACTA ACUST UNITED AC 2015; 5:95-133. [PMID: 26069080 DOI: 10.1002/9780470942390.mo140195] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Despite the dramatic increase in human lifespan over the past century, there remains pronounced variability in "health-span," or the period of time in which one is generally healthy and free of disease. Much of the variability in health-span and lifespan is thought to be genetic in origin. Understanding the genetic mechanisms of aging and identifying ways to boost longevity is a primary goal in aging research. Here, we describe a pipeline of phenotypic assays for assessing mouse models of aging. This pipeline includes behavior/cognition testing, body composition analysis, and tests of kidney function, hematopoiesis, and immune function, as well as physical parameters. We also describe study design methods for assessing lifespan and health-span, and other important considerations when conducting aging research in the laboratory mouse. The tools and assays provided can assist researchers with understanding the correlative relationships between age-associated phenotypes and, ultimately, the role of specific genes in the aging process.
Collapse
Affiliation(s)
- Cheryl L Ackert-Bicknell
- The Jackson Laboratory, Bar Harbor, Maine.,Present address: University of Rochester, Department of Orthopaedics and Rehabilitation, Rochester, New York
| | | | | | - Warren G Hill
- Laboratory of Voiding Dysfunction, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine
| | | | | | | | | |
Collapse
|
15
|
Chong J, McMillin M, Shively K, Beck A, Marvin C, Armenteros J, Buckingham K, Nkinsi N, Boyle E, Berry M, Bocian M, Foulds N, Uzielli M, Haldeman-Englert C, Hennekam R, Kaplan P, Kline A, Mercer C, Nowaczyk M, Klein Wassink-Ruiter J, McPherson E, Moreno R, Scheuerle A, Shashi V, Stevens C, Carey J, Monteil A, Lory P, Tabor H, Smith J, Shendure J, Nickerson D, Bamshad MJ, Bamshad M, Shendure J, Nickerson D, Abecasis G, Anderson P, Blue E, Annable M, Browning B, Buckingham K, Chen C, Chin J, Chong J, Cooper G, Davis C, Frazar C, Harrell T, He Z, Jain P, Jarvik G, Jimenez G, Johanson E, Jun G, Kircher M, Kolar T, Krauter S, Krumm N, Leal S, Luksic D, Marvin C, McMillin M, McGee S, O’Reilly P, Paeper B, Patterson K, Perez M, Phillips S, Pijoan J, Poel C, Reinier F, Robertson P, Santos-Cortez R, Shaffer T, Shephard C, Shively K, Siegel D, Smith J, Staples J, Tabor H, Tackett M, Underwood J, Wegener M, Wang G, Wheeler M, Yi Q. De novo mutations in NALCN cause a syndrome characterized by congenital contractures of the limbs and face, hypotonia, and developmental delay. Am J Hum Genet 2015; 96:462-73. [PMID: 25683120 DOI: 10.1016/j.ajhg.2015.01.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/07/2015] [Indexed: 12/13/2022] Open
Abstract
Freeman-Sheldon syndrome, or distal arthrogryposis type 2A (DA2A), is an autosomal-dominant condition caused by mutations in MYH3 and characterized by multiple congenital contractures of the face and limbs and normal cognitive development. We identified a subset of five individuals who had been putatively diagnosed with "DA2A with severe neurological abnormalities" and for whom congenital contractures of the limbs and face, hypotonia, and global developmental delay had resulted in early death in three cases; this is a unique condition that we now refer to as CLIFAHDD syndrome. Exome sequencing identified missense mutations in the sodium leak channel, non-selective (NALCN) in four families affected by CLIFAHDD syndrome. We used molecular-inversion probes to screen for NALCN in a cohort of 202 distal arthrogryposis (DA)-affected individuals as well as concurrent exome sequencing of six other DA-affected individuals, thus revealing NALCN mutations in ten additional families with "atypical" forms of DA. All 14 mutations were missense variants predicted to alter amino acid residues in or near the S5 and S6 pore-forming segments of NALCN, highlighting the functional importance of these segments. In vitro functional studies demonstrated that NALCN alterations nearly abolished the expression of wild-type NALCN, suggesting that alterations that cause CLIFAHDD syndrome have a dominant-negative effect. In contrast, homozygosity for mutations in other regions of NALCN has been reported in three families affected by an autosomal-recessive condition characterized mainly by hypotonia and severe intellectual disability. Accordingly, mutations in NALCN can cause either a recessive or dominant condition characterized by varied though overlapping phenotypic features, perhaps based on the type of mutation and affected protein domain(s).
Collapse
|
16
|
Bogue MA, Peters LL, Paigen B, Korstanje R, Yuan R, Ackert-Bicknell C, Grubb SC, Churchill GA, Chesler EJ. Accessing Data Resources in the Mouse Phenome Database for Genetic Analysis of Murine Life Span and Health Span. J Gerontol A Biol Sci Med Sci 2014; 71:170-7. [PMID: 25533306 PMCID: PMC4707687 DOI: 10.1093/gerona/glu223] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/28/2014] [Indexed: 01/18/2023] Open
Abstract
Understanding the source of genetic variation in aging and using this variation to define the molecular mechanisms of healthy aging require deep and broad quantification of a host of physiological, morphological, and behavioral endpoints. The murine model is a powerful system in which to understand the relations across age-related phenotypes and to identify research models with variation in life span and health span. The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging has performed broad characterization of aging in genetically diverse laboratory mice and has placed these data, along with data from several other major aging initiatives, into the interactive Mouse Phenome Database. The data may be accessed and analyzed by researchers interested in finding mouse models for specific aging processes, age-related health and disease states, and for genetic analysis of aging variation and trait covariation. We expect that by placing these data in the hands of the aging community that there will be (a) accelerated genetic analyses of aging processes, (b) discovery of genetic loci regulating life span, (c) identification of compelling correlations between life span and susceptibility for age-related disorders, and (d) discovery of concordant genomic loci influencing life span and aging phenotypes between mouse and humans.
Collapse
Affiliation(s)
- Molly A Bogue
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine.
| | - Luanne L Peters
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| | - Beverly Paigen
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| | - Ron Korstanje
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| | - Rong Yuan
- Southern Illinois University School of Medicine, Springfield
| | - Cheryl Ackert-Bicknell
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| | - Stephen C Grubb
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| | - Gary A Churchill
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| | - Elissa J Chesler
- The Jackson Laboratory Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory, Bar Harbor, Maine
| |
Collapse
|
17
|
Noordmans GA, Huang Y, Savage H, van Dijk MCRF, Schaart G, van den Bergh Weerman MA, Heeringa P, Hillebrands JL, Korstanje R, van Goor H. Genetic analysis of intracapillary glomerular lipoprotein deposits in aging mice. PLoS One 2014; 9:e111308. [PMID: 25353171 PMCID: PMC4213026 DOI: 10.1371/journal.pone.0111308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 09/30/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Renal aging is characterized by functional and structural changes like decreased glomerular filtration rate, and glomerular, tubular and interstitial damage. To gain insight in pathways involved in renal aging, we studied aged mouse strains and used genetic analysis to identify genes associated with aging phenotypes. METHODS Upon morphological screening in kidneys from 20-month-old mice from 26 inbred strains we noted intracapillary PAS-positive deposits. The severity of these deposits was quantified by scoring of a total of 50 glomeruli per section (grade 0-4). Electron microscopy and immunohistochemical staining for apoE, apoB, apoA-IV and perilipin-2 was performed to further characterize the lesions. To identify loci associated with these PAS-positive intracapillary glomerular deposits, we performed haplotype association mapping. RESULTS Six out of 26 mouse strains showed glomerular PAS-positive deposits. The severity of these deposits varied: NOD(0.97), NZW(0.41), NON(0.30), B10(0.21), C3 H(0.9) and C57BR(0.7). The intracapillary deposits were strongly positive for apoE and weakly positive for apoB and apoA-IV. Haplotype association mapping showed a strong association with a 30-Kb haplotype block on Chr 1 within the Esrrg gene. We investigated 1 Mb on each site of this region, which includes the genes Spata17, Gpatch2, Esrrg, Ush2a and Kctd3. CONCLUSIONS By analyzing 26 aged mouse strains we found that some strains developed an intracapillary PAS and apoE-positive lesion and identified a small haplotype block on Chr 1 within the Esrrg gene to be associated with these lipoprotein deposits. The region spanning this haplotype block contains the genes Spata17, Gpatch2, Esrrg, Ush2a and Kctd3, which are all highly expressed in the kidney. Esrrg might be involved in the evolvement of these glomerular deposits by influencing lipid metabolism and possibly immune reponses.
Collapse
Affiliation(s)
- Gerda A. Noordmans
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| | - Yuan Huang
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Holly Savage
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Marcory C. R. F. van Dijk
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gert Schaart
- Department of Human Movement Sciences, NUTRIM, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ron Korstanje
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Harry van Goor
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
18
|
Abstract
We sequenced the complete genome of the widely used C57L/J mouse inbred strain. With 40× average coverage, we compared the C57L/J sequence with that of the C57BL/6J and identified many known as well as novel private variants. This genome sequence adds another strain to the growing number of mouse inbred strains with complete genome sequences and is a valuable resource to the scientific community.
Collapse
|
19
|
Cochet-Bissuel M, Lory P, Monteil A. The sodium leak channel, NALCN, in health and disease. Front Cell Neurosci 2014; 8:132. [PMID: 24904279 PMCID: PMC4033012 DOI: 10.3389/fncel.2014.00132] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/28/2014] [Indexed: 12/12/2022] Open
Abstract
Ion channels are crucial components of cellular excitability and are involved in many neurological diseases. This review focuses on the sodium leak, G protein-coupled receptors (GPCRs)-activated NALCN channel that is predominantly expressed in neurons where it regulates the resting membrane potential and neuronal excitability. NALCN is part of a complex that includes not only GPCRs, but also UNC-79, UNC-80, NLF-1 and src family of Tyrosine kinases (SFKs). There is growing evidence that the NALCN channelosome critically regulates its ion conduction. Both in mammals and invertebrates, animal models revealed an involvement in many processes such as locomotor behaviors, sensitivity to volatile anesthetics, and respiratory rhythms. There is also evidence that alteration in this NALCN channelosome can cause a wide variety of diseases. Indeed, mutations in the NALCN gene were identified in Infantile Neuroaxonal Dystrophy (INAD) patients, as well as in patients with an Autosomal Recessive Syndrome with severe hypotonia, speech impairment, and cognitive delay. Deletions in NALCN gene were also reported in diseases such as 13q syndrome. In addition, genes encoding NALCN, NLF- 1, UNC-79, and UNC-80 proteins may be susceptibility loci for several diseases including bipolar disorder, schizophrenia, Alzheimer's disease, autism, epilepsy, alcoholism, cardiac diseases and cancer. Although the physiological role of the NALCN channelosome is poorly understood, its involvement in human diseases should foster interest for drug development in the near future. Toward this goal, we review here the current knowledge on the NALCN channelosome in physiology and diseases.
Collapse
Affiliation(s)
- Maud Cochet-Bissuel
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Universités Montpellier 1&2 Montpellier, France ; INSERM, U 661 Montpellier, France ; LabEx 'Ion Channel Science and Therapeutics' Montpellier, France
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Universités Montpellier 1&2 Montpellier, France ; INSERM, U 661 Montpellier, France ; LabEx 'Ion Channel Science and Therapeutics' Montpellier, France
| | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, Universités Montpellier 1&2 Montpellier, France ; INSERM, U 661 Montpellier, France ; LabEx 'Ion Channel Science and Therapeutics' Montpellier, France
| |
Collapse
|
20
|
Huang Y, Caputo CR, Noordmans GA, Yazdani S, Monteiro LH, van den Born J, van Goor H, Heeringa P, Korstanje R, Hillebrands JL. Identification of novel genes associated with renal tertiary lymphoid organ formation in aging mice. PLoS One 2014; 9:e91850. [PMID: 24637805 PMCID: PMC3956762 DOI: 10.1371/journal.pone.0091850] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 02/17/2014] [Indexed: 02/07/2023] Open
Abstract
A hallmark of aging-related organ deterioration is a dysregulated immune response characterized by pathologic leukocyte infiltration of affected tissues. Mechanisms and genes involved are as yet unknown. To identify genes associated with aging-related renal infiltration, we analyzed kidneys from aged mice (≥20 strains) for infiltrating leukocytes followed by Haplotype Association Mapping (HAM) analysis. Immunohistochemistry revealed CD45+ cell clusters (predominantly T and B cells) in perivascular areas coinciding with PNAd+ high endothelial venules and podoplanin+ lymph vessels indicative of tertiary lymphoid organs. Cumulative cluster size increased with age (analyzed at 6, 12 and 20 months). Based on the presence or absence of clusters in male and female mice at 20 months, HAM analysis revealed significant associations with loci on Chr1, Chr2, Chr8 and Chr14 in male mice, and with loci on Chr4, Chr7, Chr13 and Chr14 in female mice. Wisp2 (Chr2) showed the strongest association (P = 5.00×10−137) in male mice; Ctnnbip1 (P = 6.42×10−267) and Tnfrsf8 (P = 5.42×10−245) (both on Chr4) showed the strongest association in female mice. Both Wisp2 and Ctnnbip1 are part of the Wnt-signaling pathway and the encoded proteins were expressed within the tertiary lymphoid organs. In conclusion, this study revealed differential lymphocytic infiltration and tertiary lymphoid organ formation in aged mouse kidneys across different inbred mouse strains. HAM analysis identified candidate genes involved in the Wnt-signaling pathway that may be causally linked to tertiary lymphoid organ formation.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Pathology & Medical Biology - Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | | | - Gerda A. Noordmans
- Department of Pathology & Medical Biology - Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Saleh Yazdani
- Department of Internal Medicine - Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luiz Henrique Monteiro
- Department of Pathology & Medical Biology - Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jaap van den Born
- Department of Internal Medicine - Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology & Medical Biology - Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Heeringa
- Department of Pathology & Medical Biology - Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ron Korstanje
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Jan-Luuk Hillebrands
- Department of Pathology & Medical Biology - Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
21
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Catterall WA, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: ion channels. Br J Pharmacol 2013; 170:1607-51. [PMID: 24528239 PMCID: PMC3892289 DOI: 10.1111/bph.12447] [Citation(s) in RCA: 225] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Ion channels are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
- *
Author for correspondence;
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - William A Catterall
- University of Washington, School of Medicine, Department of PharmacologyBox 357280, Seattle, WA 98195-7280, USA
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
22
|
Noda M, Sakuta H. Central regulation of body-fluid homeostasis. Trends Neurosci 2013; 36:661-73. [DOI: 10.1016/j.tins.2013.08.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/30/2013] [Accepted: 08/12/2013] [Indexed: 01/21/2023]
|
23
|
Köroğlu Ç, Seven M, Tolun A. Recessive truncating NALCN mutation in infantile neuroaxonal dystrophy with facial dysmorphism. J Med Genet 2013; 50:515-20. [PMID: 23749988 DOI: 10.1136/jmedgenet-2013-101634] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Infantile neuroaxonal dystrophy (INAD) is a recessive disease that results in total neurological degeneration and death in childhood. PLA2G6 mutation is the underlying genetic defect, but rare genetic heterogeneity has been demonstrated. One of the five families we studied did not link to PLA2G6 locus, and in the family one of the two affected siblings additionally had atypical features including facial dysmorphism, pectus carinatum, scoliosis, pes varus, zygodactyly and bilateral cryptorchidism as well as cerebellar atrophy, as previously reported. METHODS Sural biopsy was investigated by electron microscopy. PLA2G6 was screened for mutations by Sanger sequencing. In the mutation-free family, candidate disease loci were found via linkage analysis using data from single nucleotide polymorphism genome scans. Exome sequencing was applied to find the variants at the loci. RESULTS PLA2G6 mutations were identified in four families including the one with an unusually severe phenotype that led to death within the first 2 years of life. In the remaining family, seven candidate loci totalling 15.2 Mb were found and a homozygous truncating mutation p.Q642X was identified in NALCN at 13q32.3. The patients are around 20-years-old. CONCLUSIONS NALCN is the gene responsible for INAD with facial dysmorphism. The patients have lived to adulthood despite severe growth and neuromotor retardation. NALCN forms a voltage-independent ion channel with a role in the regulation of neuronal excitability. Our findings broaden the spectrum of genes associated with neuroaxonal dystrophy. Testing infants with idiopathic severe growth retardation and neurodegeneration for NALCN mutations could benefit families.
Collapse
Affiliation(s)
- Çiğdem Köroğlu
- Department of Molecular Biology and Genetics, Boğaziçi University, Istanbul, Turkey
| | | | | |
Collapse
|
24
|
NLF-1 delivers a sodium leak channel to regulate neuronal excitability and modulate rhythmic locomotion. Neuron 2013; 77:1069-82. [PMID: 23522043 DOI: 10.1016/j.neuron.2013.01.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2013] [Indexed: 11/22/2022]
Abstract
A cation channel NCA/UNC-79/UNC-80 affects neuronal activity. We report here the identification of a conserved endoplasmic reticulum protein NLF-1 (NCA localization factor-1) that regulates neuronal excitability and locomotion through the NCA channel. In C. elegans, the loss of either NLF-1 or NCA leads to a reduced sodium leak current, and a hyperpolarized resting membrane potential in premotor interneurons. This results in a decreased premotor interneuron activity that reduces the initiation and sustainability of rhythmic locomotion. NLF-1 promotes axonal localization of all NCA reporters. Its mouse homolog mNLF-1 functionally substitutes for NLF-1 in C. elegans, interacts with the mammalian sodium leak channel NALCN in vitro, and potentiates sodium leak currents in primary cortical neuron cultures. Taken together, an ER protein NLF-1 delivers a sodium leak channel to maintain neuronal excitability and potentiates a premotor interneuron network critical for C. elegans rhythmic locomotion.
Collapse
|
25
|
Senatore A, Spafford JD. A uniquely adaptable pore is consistent with NALCN being an ion sensor. Channels (Austin) 2013; 7:60-8. [PMID: 23442378 PMCID: PMC3667885 DOI: 10.4161/chan.23981] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
NALCN is an intriguing, orphan ion channel among the 4x6TM family of related voltage-gated cation channels, sharing a common architecture of four homologous domains consisting of six transmembrane helices, separated by three cytoplasmic linkers and delimited by N and C-terminal ends. NALCN is one of the shortest 4x6TM family members, lacking much of the variation that provides the diverse palate of gating features, and tissue specific adaptations of sodium and calcium channels. NALCN's most distinctive feature is that that it possesses a highly adaptable pore with a calcium-like EEEE selectivity filter in radially symmetrical animals and a more sodium-like EEKE or EKEE selectivity filter in bilaterally symmetrical animals including vertebrates. Two lineages of animals evolved alternative calcium-like EEEE and sodium-like EEKE / EKEE pores, spliced to regulate NALCN functions in differing cellular environments, such as muscle (heart and skeletal) and secretory tissue (brain and glands), respectively. A highly adaptable pore in an otherwise conserved ion channel in the 4x6TM channel family is not consistent with a role for NALCN in directly gating a significant ion conductance that can be either sodium ions or calcium ions. NALCN was proposed to be an expressible Gd ( 3+) -sensitive, NMDG (+) -impermeant, non-selective and ohmic leak conductance in HEK-293T cells, but we were unable to distinguish these reported currents from leaky patch currents (ILP) in control HEK-293T cells. We suggest that NALCN functions as a sensor for the much larger UNC80/UNC79 complex, in a manner consistent with the coupling mechanism known for other weakly or non-conducting 4x6TM channel sensor proteins such as Nax or Cav 1.1. We propose that NALCN serves as a variable sensor that responds to calcium or sodium ion flux, depending on whether the total cellular current density is generated more from calcium-selective or sodium-selective channels.
Collapse
Affiliation(s)
- Adriano Senatore
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | | |
Collapse
|
26
|
Senatore A, Monteil A, van Minnen J, Smit AB, Spafford JD. NALCN ion channels have alternative selectivity filters resembling calcium channels or sodium channels. PLoS One 2013; 8:e55088. [PMID: 23383067 PMCID: PMC3557258 DOI: 10.1371/journal.pone.0055088] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 12/19/2012] [Indexed: 11/17/2022] Open
Abstract
NALCN is a member of the family of ion channels with four homologous, repeat domains that include voltage-gated calcium and sodium channels. NALCN is a highly conserved gene from simple, extant multicellular organisms without nervous systems such as sponges and placozoans and mostly remains a single gene compared to the calcium and sodium channels which diversified into twenty genes in humans. The single NALCN gene has alternatively-spliced exons at exons 15 or exon 31 that splices in novel selectivity filter residues that resemble calcium channels (EEEE) or sodium channels (EKEE or EEKE). NALCN channels with alternative calcium, (EEEE) and sodium, (EKEE or EEKE) -selective pores are conserved in simple bilaterally symmetrical animals like flatworms to non-chordate deuterostomes. The single NALCN gene is limited as a sodium channel with a lysine (K)-containing pore in vertebrates, but originally NALCN was a calcium-like channel, and evolved to operate as both a calcium channel and sodium channel for different roles in many invertebrates. Expression patterns of NALCN-EKEE in pond snail, Lymnaea stagnalis suggest roles for NALCN in secretion, with an abundant expression in brain, and an up-regulation in secretory organs of sexually-mature adults such as albumen gland and prostate. NALCN-EEEE is equally abundant as NALCN-EKEE in snails, but is greater expressed in heart and other muscle tissue, and 50% less expressed in the brain than NALCN-EKEE. Transfected snail NALCN-EEEE and NALCN-EKEE channel isoforms express in HEK-293T cells. We were not able to distinguish potential NALCN currents from background, non-selective leak conductances in HEK293T cells. Native leak currents without expressing NALCN genes in HEK-293T cells are NMDG(+) impermeant and blockable with 10 µM Gd(3+) ions and are indistinguishable from the hallmark currents ascribed to mammalian NALCN currents expressed in vitro by Lu et al. in Cell. 2007 Apr 20;129(2):371-83.
Collapse
|
27
|
Genomic and genome-wide association of susceptibility to radiation-induced fibrotic lung disease in mice. Radiother Oncol 2012; 105:350-7. [DOI: 10.1016/j.radonc.2012.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 07/10/2012] [Accepted: 08/11/2012] [Indexed: 12/13/2022]
|
28
|
Lu TZ, Feng ZP. NALCN: A Regulator of Pacemaker Activity. Mol Neurobiol 2012; 45:415-23. [DOI: 10.1007/s12035-012-8260-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/09/2012] [Indexed: 11/25/2022]
|
29
|
Vantyghem MC, Balavoine AS, Wémeau JL, Douillard C. Hyponatremia and antidiuresis syndrome. ANNALES D'ENDOCRINOLOGIE 2011; 72:500-12. [PMID: 22119069 DOI: 10.1016/j.ando.2011.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 10/07/2011] [Indexed: 12/27/2022]
Abstract
Antidiuretic hormone (ADH), or arginine vasopressin (AVP), is primarily regulated through plasma osmolarity, as well as non-osmotic stimuli including blood volume and stress. Links between water-electrolyte and carbohydrate metabolism have also been recently demonstrated. AVP acts via the intermediary of three types of receptors: V1a, or V1, which exerts vasoconstrictive effects; pituitary gland V1b, or V3, which participates in the secretion of ACTH; and renal V2, which reduces the excretion of pure water by combining with water channels (aquaporin 2). Antidiuresis syndrome is a form of euvolaemic, hypoosmolar hyponatraemia, which is characterised by a negative free water clearance with inappropriate urine osmolality and intracellular hyper-hydration in the absence of renal, adrenal and thyroid insufficiency. Ninety percent of cases of antidiuresis syndrome occur in association with hypersecretion of vasopressin, while vasopressin is undetectable in 10% of cases. Thus the term "antidiuresis syndrome" is more appropriate than the classic name "syndrome of inappropriate ADH secretion" (SIADH). The clinical symptoms, morbidity and mortality of hyponatraemia are related to its severity, as well as to the rapidity of its onset and duration. Even in cases of moderate hyponatraemia that are considered asymptomatic, there is a very high risk of falls due to gait and attention disorders, as well as rhabdomyolysis, which increases the fracture risk. The aetiological diagnosis of hyponatraemia is based on the analysis of calculated or measured plasma osmolality (POsm), as well as blood volume (skin tenting of dehydration, oedema). Hyperglycaemia and hypertriglyceridaemia lead to hyper- and normoosmolar hyponatraemia, respectively. Salt loss of gastrointestinal, renal, cutaneous and sometimes cerebral origin is hypovolaemic, hypoosmolar hyponatraemia (skin tenting), whereas oedema is present with hypervolaemic, hypoosmolar hyponatraemia of heart failure, nephrotic syndrome and cirrhosis. Some endocrinopathies (glucocorticoid deficiency and hypothyroidism) are associated with euvolaemic, hypoosmolar hyponatraemia, which must be distinguished from SIADH. Independent of adrenal insufficiency, isolated hypoaldosteronism can also be accompanied by hypersecretion of vasopressin secondary to hypovolaemia, which responds to mineralocorticoid administration. The causes of SIADH are classic: neoplastic (notably small-cell lung cancer), iatrogenic (particularly psychoactive drugs, chemotherapy), lung and cerebral. Some causes have been recently described: familial hyponatraemia via X-linked recessive disease caused by an activating mutation of the vasopressin 2 receptor; and corticotropin insufficiency related to drug interference between some inhaled glucocorticoids and cytochrome p450 inhibitors, such as the antiretroviral drugs and itraconazole, etc. SIADH in marathon runners exposes them to a risk of hypotonic encephalopathy with fatal cerebral oedema. SIADH treatment is based on water restriction and demeclocycline. V2 receptor antagonists are still not marketed in France. These aquaretics seem effective clinically and biologically, without demonstrated improvement to date of mortality in eu- and hypervolaemic hyponatraemia. Obviously treatment of a corticotropic deficit, even subtle, should not be overlooked, as well as the introduction of fludrocortisone in isolated hypoaldosteronism and discontinuation of iatrogenic drugs.
Collapse
Affiliation(s)
- Marie-Christine Vantyghem
- Service d'endocrinologie et maladies métaboliques, hôpital Huriez, centre hospitalier régional universitaire de Lille, 1, rue Polonovski, 59000 Lille, France.
| | | | | | | |
Collapse
|
30
|
Sinke AP, Deen PMT. The physiological implication of novel proteins in systemic osmoregulation. FASEB J 2011; 25:3279-89. [PMID: 21737616 DOI: 10.1096/fj.11-188433] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Maintenance of the osmobalance is important for life. In this process, in which brain and kidney act in concert, mammals have to cope with significant deviations as drinking water reduces plasma osmolality, whereas salty food increases it. To restore homeostasis, specialized nuclei within the hypothalamus play a pivotal role in detecting changes in plasma osmolality and initiating appropriate responses. These responses are accomplished by either changing the intake of water or the excretion of water by the kidney. In the past decade, several novel findings have made significant contributions to our insights in the process of systemic osmoregulation. Novel proteins have been identified in the brain as well as in the kidney that are fulfilling important roles in the process of systemic osmoregulation. In this review, recent evidence of the involvement of TRPV channels (TRPV1, TRPV2, and TRPV4) and proteins, such as sodium channels NALCN and Na(x), in neuronal osmoregulation, as well as; e.g., the purinergic P2Y2 receptor in renal osmoregulation, are discussed, and integrated with existing knowledge of systemic osmoregulation.
Collapse
Affiliation(s)
- Anne P Sinke
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|