78 results on '"Rajan Sah"'
Search Results
2. Regulation of the Volume-Regulated Anion Channel Pore-Forming Subunit LRRC8A in the Intrahippocampal Kainic Acid Model of Mesial Temporal Lobe Epilepsy
- Author
-
Manolia R. Ghouli, Carrie R. Jonak, Rajan Sah, Todd A. Fiacco, and Devin K. Binder
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Volume-regulated anion channels (VRACs) are a group of ubiquitously expressed outwardly-rectifying anion channels that sense increases in cell volume and act to return cells to baseline volume through an efflux of anions and organic osmolytes, including glutamate. Because cell swelling, increased extracellular glutamate levels, and reduction of the brain extracellular space (ECS) all occur during seizure generation, we set out to determine whether VRACs are dysregulated throughout mesial temporal lobe epilepsy (MTLE), the most common form of adult epilepsy. To accomplish this, we employed the IHKA experimental model of MTLE, and probed for the expression of LRRC8A, the essential pore-forming VRAC subunit, at acute, early-, mid-, and late-epileptogenic time points (1-, 7-, 14-, and 30-days post-IHKA, respectively). Western blot analysis revealed the upregulation of total dorsal hippocampal LRRC8A 14-days post-IHKA in both the ipsilateral and contralateral hippocampus. Immunohistochemical analyses showed an increased LRRC8A signal 7-days post-IHKA in both the ipsilateral and contralateral hippocampus, along with layer-specific changes 1-, 7-, and 30-days post-IHKA bilaterally. LRRC8A upregulation 1 day post-IHKA was observed primarily in astrocytes; however, some upregulation was also observed in neurons. Glutamate-GABA/glutamine cycle enzymes glutamic acid decarboxylase, glutaminase, and glutamine synthetase were also dysregulated at the 7-day timepoint post status epilepticus. The timepoint-dependent upregulation of total hippocampal LRRC8A and the possible subsequent increased efflux of glutamate in the epileptic hippocampus suggest that the dysregulation of astrocytic VRAC may play an important role in the development of epilepsy.
- Published
- 2023
- Full Text
- View/download PDF
3. Conditional deletion of LRRC8A in the brain reduces stroke damage independently of swelling-activated glutamate release
- Author
-
Mustafa Balkaya, Preeti Dohare, Sophie Chen, Alexandra L. Schober, Antonio M. Fidaleo, Julia W. Nalwalk, Rajan Sah, and Alexander A. Mongin
- Subjects
Molecular biology ,Molecular neuroscience ,Cell biology ,Science - Abstract
Summary: The ubiquitous volume-regulated anion channels (VRACs) facilitate cell volume control and contribute to many other physiological processes. Treatment with non-specific VRAC blockers or brain-specific deletion of the essential VRAC subunit LRRC8A is highly protective in rodent models of stroke. Here, we tested the widely accepted idea that the harmful effects of VRACs are mediated by release of the excitatory neurotransmitter glutamate. We produced conditional LRRC8A knockout either exclusively in astrocytes or in the majority of brain cells. Genetically modified mice were subjected to an experimental stroke (middle cerebral artery occlusion). The astrocytic LRRC8A knockout yielded no protection. Conversely, the brain-wide LRRC8A deletion strongly reduced cerebral infarction in both heterozygous (Het) and full KO mice. Yet, despite identical protection, Het mice had full swelling-activated glutamate release, whereas KO animals showed its virtual absence. These findings suggest that LRRC8A contributes to ischemic brain injury via a mechanism other than VRAC-mediated glutamate release.
- Published
- 2023
- Full Text
- View/download PDF
4. Adipose-targeted SWELL1 deletion exacerbates obesity- and age-related nonalcoholic fatty liver disease
- Author
-
Susheel K. Gunasekar, John Heebink, Danielle H. Carpenter, Ashutosh Kumar, Litao Xie, Haixia Zhang, Joel D. Schilling, and Rajan Sah
- Subjects
Hepatology ,Metabolism ,Medicine - Abstract
Healthy expansion of adipose tissue is critical for the maintenance of metabolic health, providing an optimized reservoir for energy storage in the form of triacylglycerol-rich lipoproteins. Dysfunctional adipocytes that are unable to efficiently store lipid can result in lipodystrophy and contribute to nonalcoholic fatty liver disease (NAFLD) and metabolic syndrome. Leucine-rich repeat containing protein 8a/SWELL1 functionally encodes the volume-regulated anion channel complex in adipocytes, is induced in early obesity, and is required for normal adipocyte expansion during high-fat feeding. Adipose-specific SWELL1 ablation (Adipo KO) leads to insulin resistance and hyperglycemia during caloric excess, both of which are associated with NAFLD. Here, we show that Adipo-KO mice exhibited impaired adipose depot expansion and excess lipolysis when raised on a variety of high-fat diets, resulting in increased diacylglycerides and hepatic steatosis, thereby driving liver injury. Liver lipidomic analysis revealed increases in oleic acid–containing hepatic triacylglycerides and injurious hepatic diacylglyceride species, with reductions in hepatocyte-protective phospholipids and antiinflammatory free fatty acids. Aged Adipo-KO mice developed hepatic steatosis on a regular chow diet, and Adipo-KO male mice developed spontaneous, aggressive hepatocellular carcinomas (HCCs). These data highlight the importance of adipocyte SWELL1 for healthy adipocyte expansion to protect against NAFLD and HCC in the setting of overnutrition and with aging.
- Published
- 2023
- Full Text
- View/download PDF
5. Small molecule SWELL1 complex induction improves glycemic control and nonalcoholic fatty liver disease in murine Type 2 diabetes
- Author
-
Susheel K. Gunasekar, Litao Xie, Ashutosh Kumar, Juan Hong, Pratik R. Chheda, Chen Kang, David M. Kern, Chau My-Ta, Joshua Maurer, John Heebink, Eva E. Gerber, Wojciech J. Grzesik, Macaulay Elliot-Hudson, Yanhui Zhang, Phillip Key, Chaitanya A. Kulkarni, Joseph W. Beals, Gordon I. Smith, Isaac Samuel, Jessica K. Smith, Peter Nau, Yumi Imai, Ryan D. Sheldon, Eric B. Taylor, Daniel J. Lerner, Andrew W. Norris, Samuel Klein, Stephen G. Brohawn, Robert Kerns, and Rajan Sah
- Subjects
Science - Abstract
Type 2 diabetes is associated with insulin resistance, impaired insulin secretion and liver steatosis. Here the authors report a proof-of-concept study for small molecule SWELL1 modulators as a therapeutic approach to treat diabetes and associated liver steatosis by enhancing systemic insulin-sensitivity and insulin secretion in mice.
- Published
- 2022
- Full Text
- View/download PDF
6. Electrophysiological and Molecular Mechanisms of Sinoatrial Node Mechanosensitivity
- Author
-
Daniel Turner, Chen Kang, Pietro Mesirca, Juan Hong, Matteo E. Mangoni, Alexey V. Glukhov, and Rajan Sah
- Subjects
automaticity ,ion channel ,cardiac ,stretch activated ,calcium ,heart rate ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
The understanding of the electrophysiological mechanisms that underlie mechanosensitivity of the sinoatrial node (SAN), the primary pacemaker of the heart, has been evolving over the past century. The heart is constantly exposed to a dynamic mechanical environment; as such, the SAN has numerous canonical and emerging mechanosensitive ion channels and signaling pathways that govern its ability to respond to both fast (within second or on beat-to-beat manner) and slow (minutes) timescales. This review summarizes the effects of mechanical loading on the SAN activity and reviews putative candidates, including fast mechanoactivated channels (Piezo, TREK, and BK) and slow mechanoresponsive ion channels [including volume-regulated chloride channels and transient receptor potential (TRP)], as well as the components of mechanochemical signal transduction, which may contribute to SAN mechanosensitivity. Furthermore, we examine the structural foundation for both mechano-electrical and mechanochemical signal transduction and discuss the role of specialized membrane nanodomains, namely, caveolae, in mechanical regulation of both membrane and calcium clock components of the so-called coupled-clock pacemaker system responsible for SAN automaticity. Finally, we emphasize how these mechanically activated changes contribute to the pathophysiology of SAN dysfunction and discuss controversial areas necessitating future investigations. Though the exact mechanisms of SAN mechanosensitivity are currently unknown, identification of such components, their impact into SAN pacemaking, and pathological remodeling may provide new therapeutic targets for the treatment of SAN dysfunction and associated rhythm abnormalities.
- Published
- 2021
- Full Text
- View/download PDF
7. SWELL signalling in adipocytes: can fat 'feel' fat?
- Author
-
Susheel K. Gunasekar, Litao Xie, and Rajan Sah
- Subjects
obesity ,ion channel ,insulin ,caveolae ,mechano-transduction ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 ,Cytology ,QH573-671 ,Physiology ,QP1-981 - Abstract
Obesity is becoming a global epidemic, predisposing to Type 2 diabetes, cardiovascular disease, fatty liver disease, pulmonary disease, osteoarthritis and cancer. Therefore, understanding the biology of adipocyte expansion in response to overnutrition is critical to devising strategies to treat obesity, and the associated burden of morbidity and mortality. Through exploratory patch-clamp experiments in freshly isolated primary murine and human adipocytes, we recently determined that SWELL1/LRRC8a, a leucine-rich repeat containing transmembrane protein, functionally encoded an ion channel signalling complex (the volume-regulated anion channel, or VRAC) on the adipocyte plasma membrane. The SWELL1-/LRRC8 channel complex activates in response to increases in adipocyte volume and in the context of obesity. SWELL1 is also required for insulin-PI3K-AKT2 signalling to regulate adipocyte growth and systemic glycaemia. This commentary delves further into our working models for the molecular mechanisms of adipocyte SWELL1-mediated VRAC activation, proposed signal transduction mechanisms, and putative impact on adipocyte hypertrophy during caloric excess.
- Published
- 2019
- Full Text
- View/download PDF
8. Perilipin 2 downregulation in β cells impairs insulin secretion under nutritional stress and damages mitochondria
- Author
-
Akansha Mishra, Siming Liu, Joseph Promes, Mikako Harata, William Sivitz, Brian Fink, Gourav Bhardwaj, Brian T. O’Neill, Chen Kang, Rajan Sah, Stefan Strack, Samuel Stephens, Timothy King, Laura Jackson, Andrew S. Greenberg, Frederick Anokye-Danso, Rexford S. Ahima, James Ankrum, and Yumi Imai
- Subjects
Endocrinology ,Metabolism ,Medicine - Abstract
Perilipin 2 (PLIN2) is a lipid droplet (LD) protein in β cells that increases under nutritional stress. Downregulation of PLIN2 is often sufficient to reduce LD accumulation. To determine whether PLIN2 positively or negatively affects β cell function under nutritional stress, PLIN2 was downregulated in mouse β cells, INS1 cells, and human islet cells. β Cell–specific deletion of PLIN2 in mice on a high-fat diet reduced glucose-stimulated insulin secretion (GSIS) in vivo and in vitro. Downregulation of PLIN2 in INS1 cells blunted GSIS after 24-hour incubation with 0.2 mM palmitic acid. Downregulation of PLIN2 in human pseudoislets cultured at 5.6 mM glucose impaired both phases of GSIS, indicating that PLIN2 is critical for GSIS. Downregulation of PLIN2 decreased specific OXPHOS proteins in all 3 models and reduced oxygen consumption rates in INS1 cells and mouse islets. Moreover, we found that PLIN2-deficient INS1 cells increased the distribution of a fluorescent oleic acid analog to mitochondria and showed signs of mitochondrial stress, as indicated by susceptibility to fragmentation and alterations of acyl-carnitines and glucose metabolites. Collectively, PLIN2 in β cells has an important role in preserving insulin secretion, β cell metabolism, and mitochondrial function under nutritional stress.
- Published
- 2021
- Full Text
- View/download PDF
9. The SWELL1-LRRC8 complex regulates endothelial AKT-eNOS signaling and vascular function
- Author
-
Ahmad F Alghanem, Javier Abello, Joshua M Maurer, Ashutosh Kumar, Chau My Ta, Susheel K Gunasekar, Urooj Fatima, Chen Kang, Litao Xie, Oluwaseun Adeola, Megan Riker, Macaulay Elliot-Hudson, Rachel A Minerath, Chad E Grueter, Robert F Mullins, Amber N Stratman, and Rajan Sah
- Subjects
ion channel ,mechanobiology ,hypertension ,diabetes ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The endothelium responds to numerous chemical and mechanical factors in regulating vascular tone, blood pressure, and blood flow. The endothelial volume-regulated anion channel (VRAC) has been proposed to be mechanosensitive and thereby sense fluid flow and hydrostatic pressure to regulate vascular function. Here, we show that the leucine-rich repeat-containing protein 8a, LRRC8A (SWELL1), is required for VRAC in human umbilical vein endothelial cells (HUVECs). Endothelial LRRC8A regulates AKT-endothelial nitric oxide synthase (eNOS) signaling under basal, stretch, and shear-flow stimulation, forms a GRB2-Cav1-eNOS signaling complex, and is required for endothelial cell alignment to laminar shear flow. Endothelium-restricted Lrrc8a KO mice develop hypertension in response to chronic angiotensin-II infusion and exhibit impaired retinal blood flow with both diffuse and focal blood vessel narrowing in the setting of type 2 diabetes (T2D). These data demonstrate that LRRC8A regulates AKT-eNOS in endothelium and is required for maintaining vascular function, particularly in the setting of T2D.
- Published
- 2021
- Full Text
- View/download PDF
10. SWELL1 regulates skeletal muscle cell size, intracellular signaling, adiposity and glucose metabolism
- Author
-
Ashutosh Kumar, Litao Xie, Chau My Ta, Antentor O Hinton, Susheel K Gunasekar, Rachel A Minerath, Karen Shen, Joshua M Maurer, Chad E Grueter, E Dale Abel, Gretchen Meyer, and Rajan Sah
- Subjects
VRAC ,ion channel ,knock-out mouse ,growth ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Maintenance of skeletal muscle is beneficial in obesity and Type 2 diabetes. Mechanical stimulation can regulate skeletal muscle differentiation, growth and metabolism; however, the molecular mechanosensor remains unknown. Here, we show that SWELL1 (Lrrc8a) functionally encodes a swell-activated anion channel that regulates PI3K-AKT, ERK1/2, mTOR signaling, muscle differentiation, myoblast fusion, cellular oxygen consumption, and glycolysis in skeletal muscle cells. LRRC8A over-expression in Lrrc8a KO myotubes boosts PI3K-AKT-mTOR signaling to supra-normal levels and fully rescues myotube formation. Skeletal muscle-targeted Lrrc8a KO mice have smaller myofibers, generate less force ex vivo, and exhibit reduced exercise endurance, associated with increased adiposity under basal conditions, and glucose intolerance and insulin resistance when raised on a high-fat diet, compared to wild-type (WT) mice. These results reveal that the LRRC8 complex regulates insulin-PI3K-AKT-mTOR signaling in skeletal muscle to influence skeletal muscle differentiation in vitro and skeletal myofiber size, muscle function, adiposity and systemic metabolism in vivo.
- Published
- 2020
- Full Text
- View/download PDF
11. SWELL1 is a glucose sensor regulating β-cell excitability and systemic glycaemia
- Author
-
Chen Kang, Litao Xie, Susheel K. Gunasekar, Anil Mishra, Yanhui Zhang, Saachi Pai, Yiwen Gao, Ashutosh Kumar, Andrew W. Norris, Samuel B. Stephens, and Rajan Sah
- Subjects
Science - Abstract
Insulin secretion by β-cells is stimulated by glucose and is dependent on the induction of β-cell membrane depolarization, mainly driven by the closure of KATP channels, which in turn promotes voltage-gated Ca2+ channel opening. Here Kang et al. show that the volume-regulated anion channel, SWELL1, is involved in glucose-stimulated calcium increase and insulin secretion.
- Published
- 2018
- Full Text
- View/download PDF
12. Transient Receptor Potential channels (TRP) in GtoPdb v.2023.1
- Author
-
Michael X. Zhu, Lixia Yue, Wei Yang, Fan Yang, Haoxing Xu, Long-Jun Wu, Joris Vriens, Dan Tong, Jinbin Tian, Stephanie C. Stotz, Rajan Sah, Antonio Riccio, Grzegorz Owsianik, Elena Oancea, Bernd Nilius, David McKemy, Qiang Liu, Boyi Liu, Kristopher T Kahle, David Julius, Sven E. Jordt, Meiqin Hu, Kotdaji Ha, Christian M. Grimm, Lu Fan, Julia F. Doerner, Markus Delling, Katrien De Clerq, David E. Clapham, Dipayan Chaudhuri, Ingrid Carvacho, and Nathaniel T. Blair
- Subjects
General Medicine ,General Chemistry - Abstract
The TRP superfamily of channels (nomenclature as agreed by NC-IUPHAR [176, 1072]), whose founder member is the Drosophila Trp channel, exists in mammals as six families; TRPC, TRPM, TRPV, TRPA, TRPP and TRPML based on amino acid homologies. TRP subunits contain six putative TM domains and assemble as homo- or hetero-tetramers to form cation selective channels with diverse modes of activation and varied permeation properties (reviewed by [730]). Established, or potential, physiological functions of the individual members of the TRP families are discussed in detail in the recommended reviews and in a number of books [401, 686, 1155, 256]. The established, or potential, involvement of TRP channels in disease [1126] is reviewed in [448, 685], [688] and [464], together with a special edition of Biochemica et Biophysica Acta on the subject [685]. Additional disease related reviews, for pain [633], stroke [1135], sensation and inflammation [988], itch [130], and airway disease [310, 1051], are available. The pharmacology of most TRP channels has been advanced in recent years. Broad spectrum agents are listed in the tables along with more selective, or recently recognised, ligands that are flagged by the inclusion of a primary reference. See Rubaiy (2019) for a review of pharmacological tools for TRPC1/C4/C5 channels [805]. Most TRP channels are regulated by phosphoinostides such as PtIns(4,5)P2 although the effects reported are often complex, occasionally contradictory, and likely to be dependent upon experimental conditions, such as intracellular ATP levels (reviewed by [1009, 689, 801]). Such regulation is generally not included in the tables.When thermosensitivity is mentioned, it refers specifically to a high Q10 of gating, often in the range of 10-30, but does not necessarily imply that the channel's function is to act as a 'hot' or 'cold' sensor. In general, the search for TRP activators has led to many claims for temperature sensing, mechanosensation, and lipid sensing. All proteins are of course sensitive to energies of binding, mechanical force, and temperature, but the issue is whether the proposed input is within a physiologically relevant range resulting in a response. TRPA (ankyrin) familyTRPA1 is the sole mammalian member of this group (reviewed by [293]). TRPA1 activation of sensory neurons contribute to nociception [414, 890, 602]. Pungent chemicals such as mustard oil (AITC), allicin, and cinnamaldehyde activate TRPA1 by modification of free thiol groups of cysteine side chains, especially those located in its amino terminus [575, 60, 365, 577]. Alkenals with α, β-unsaturated bonds, such as propenal (acrolein), butenal (crotylaldehyde), and 2-pentenal can react with free thiols via Michael addition and can activate TRPA1. However, potency appears to weaken as carbon chain length increases [26, 60]. Covalent modification leads to sustained activation of TRPA1. Chemicals including carvacrol, menthol, and local anesthetics reversibly activate TRPA1 by non-covalent binding [424, 511, 1081, 1080]. TRPA1 is not mechanosensitive under physiological conditions, but can be activated by cold temperatures [425, 212]. The electron cryo-EM structure of TRPA1 [740] indicates that it is a 6-TM homotetramer. Each subunit of the channel contains two short ‘pore helices’ pointing into the ion selectivity filter, which is big enough to allow permeation of partially hydrated Ca2+ ions. TRPC (canonical) familyMembers of the TRPC subfamily (reviewed by [284, 778, 18, 4, 94, 446, 739, 70]) fall into the subgroups outlined below. TRPC2 is a pseudogene in humans. It is generally accepted that all TRPC channels are activated downstream of Gq/11-coupled receptors, or receptor tyrosine kinases (reviewed by [765, 953, 1072]). A comprehensive listing of G-protein coupled receptors that activate TRPC channels is given in [4]. Hetero-oligomeric complexes of TRPC channels and their association with proteins to form signalling complexes are detailed in [18] and [447]. TRPC channels have frequently been proposed to act as store-operated channels (SOCs) (or compenents of mulimeric complexes that form SOCs), activated by depletion of intracellular calcium stores (reviewed by [741, 18, 770, 820, 1121, 157, 726, 64, 158]). However, the weight of the evidence is that they are not directly gated by conventional store-operated mechanisms, as established for Stim-gated Orai channels. TRPC channels are not mechanically gated in physiologically relevant ranges of force. All members of the TRPC family are blocked by 2-APB and SKF96365 [347, 346]. Activation of TRPC channels by lipids is discussed by [70]. Important progress has been recently made in TRPC pharmacology [805, 619, 436, 102, 851, 191, 291]. TRPC channels regulate a variety of physiological functions and are implicated in many human diseases [295, 71, 885, 1031, 1025, 154, 103, 561, 913, 409]. TRPC1/C4/C5 subgroup TRPC1 alone may not form a functional ion channel [229]. TRPC4/C5 may be distinguished from other TRP channels by their potentiation by micromolar concentrations of La3+. TRPC2 is a pseudogene in humans, but in other mammals appears to be an ion channel localized to microvilli of the vomeronasal organ. It is required for normal sexual behavior in response to pheromones in mice. It may also function in the main olfactory epithelia in mice [1114, 723, 724, 1115, 539, 1168, 1109].TRPC3/C6/C7 subgroup All members are activated by diacylglycerol independent of protein kinase C stimulation [347].TRPM (melastatin) familyMembers of the TRPM subfamily (reviewed by [275, 346, 741, 1151]) fall into the five subgroups outlined below. TRPM1/M3 subgroupIn darkness, glutamate released by the photoreceptors and ON-bipolar cells binds to the metabotropic glutamate receptor 6 , leading to activation of Go . This results in the closure of TRPM1. When the photoreceptors are stimulated by light, glutamate release is reduced, and TRPM1 channels are more active, resulting in cell membrane depolarization. Human TRPM1 mutations are associated with congenital stationary night blindness (CSNB), whose patients lack rod function. TRPM1 is also found melanocytes. Isoforms of TRPM1 may present in melanocytes, melanoma, brain, and retina. In melanoma cells, TRPM1 is prevalent in highly dynamic intracellular vesicular structures [398, 708]. TRPM3 (reviewed by [714]) exists as multiple splice variants which differ significantly in their biophysical properties. TRPM3 is expressed in somatosensory neurons and may be important in development of heat hyperalgesia during inflammation (see review [941]). TRPM3 is frequently coexpressed with TRPA1 and TRPV1 in these neurons. TRPM3 is expressed in pancreatic beta cells as well as brain, pituitary gland, eye, kidney, and adipose tissue [713, 940]. TRPM3 may contribute to the detection of noxious heat [1017]. TRPM2TRPM2 is activated under conditions of oxidative stress (respiratory burst of phagocytic cells). The direct activators are calcium, adenosine diphosphate ribose (ADPR) [970] and cyclic ADPR (cADPR) [1118]. As for many ion channels, PI(4,5)P2 must also be present [1109]. Numerous splice variants of TRPM2 exist which differ in their activation mechanisms [239]. Recent studies have reported structures of human (hs) TRPM2, which demonstrate two ADPR binding sites in hsTRPM2, one in the N-terminal MHR1/2 domain and the other in the C-terminal NUDT9-H domain. In addition, one Ca2+ binding site in the intracellular S2-S3 loop is revealed and proposed to mediate Ca2+ binding that induces conformational changes leading the ADPR-bound closed channel to open [387, 1027]. Meanwhile, a quadruple-residue motif (979FGQI982) was identified as the ion selectivity filter and a gate to control ion permeation in hsTRPM2 [1120]. TRPM2 is involved in warmth sensation [848], and contributes to several diseases [76]. TRPM2 interacts with extra synaptic NMDA receptors (NMDAR) and enhances NMDAR activity in ischemic stroke [1164]. Activation of TRPM2 in macrophages promotes atherosclerosis [1165, 1147]. Moreover, silica nanoparticles induce lung inflammation in mice via ROS/PARP/TRPM2 signaling-mediated lysosome impairment and autophagy dysfunction [1028]. Recent studies have designed various compounds for their potential to selectively inhibit the TRPM2 channel, including ACA derivatives A23, and 2,3-dihydroquinazolin-4(1H)-one derivatives [1137, 1139]. TRPM4/5 subgroupTRPM4 and TRPM5 have the distinction within all TRP channels of being impermeable to Ca2+ [1072]. A splice variant of TRPM4 (i.e.TRPM4b) and TRPM5 are molecular candidates for endogenous calcium-activated cation (CAN) channels [327]. TRPM4 is active in the late phase of repolarization of the cardiac ventricular action potential. TRPM4 deletion or knockout enhances beta adrenergic-mediated inotropy [593]. Mutations are associated with conduction defects [404, 593, 879]. TRPM4 has been shown to be an important regulator of Ca2+ entry in to mast cells [993] and dendritic cell migration [52]. TRPM5 in taste receptor cells of the tongue appears essential for the transduction of sweet, amino acid and bitter stimuli [537] TRPM5 contributes to the slow afterdepolarization of layer 5 neurons in mouse prefrontal cortex [513]. Both TRPM4 and TRPM5 are required transduction of taste stimuli [246]. TRPM6/7 subgroupTRPM6 and 7 combine channel and enzymatic activities (‘chanzymes’) [172]. These channels have the unusual property of permeation by divalent (Ca2+, Mg2+, Zn2+) and monovalent cations, high single channel conductances, but overall extremely small inward conductance when expressed to the plasma membrane. They are inhibited by internal Mg2+ at ~0.6 mM, around the free level of Mg2+ in cells. Whether they contribute to Mg2+ homeostasis is a contentious issue. PIP2 is required for TRPM6 and TRPM7 activation [810, 1077]. When either gene is deleted in mice, the result is embryonic lethality [413, 1065]. The C-terminal kinase region of TRPM6 and TRPM7 is cleaved under unknown stimuli, and the kinase phosphorylates nuclear histones [479, 480]. TRPM7 is responsible for oxidant- induced Zn2+ release from intracellular vesicles [3] and contributes to intestinal mineral absorption essential for postnatal survival [622]. The putative metal transporter proteins CNNM1-4 interact with TRPM7 and regulate TRPM7 channel activity [40, 467]. TRPM8Is a channel activated by cooling and pharmacological agents evoking a ‘cool’ sensation and participates in the thermosensation of cold temperatures [63, 178, 224] reviewed by [1011, 562, 457, 649]. Direct chemical agonists include menthol and icilin[1086]. Besides, linalool can promote ERK phosphorylation in human dermal microvascular endothelial cells, down-regulate intracellular ATP levels, and activate TRPM8 [68]. Recent studies have found that TRPM8 has typical S4-S5 connectomes with clear selective filters and exowell rings [512], and have identified cryo-electron microscopy structures of mouse TRPM8 in closed, intermediate, and open states along the ligand- and PIP2-dependent gated pathways [1111]. Moreover, the last 36 amino acids at the carboxyl terminal of TRPM8 are key protein sequences for TRPM8's temperature-sensitive function [194]. TRPM8 deficiency reduced the expression of S100A9 and increased the expression of HNF4α in the liver of mice, which reduced inflammation and fibrosis progression in mice with liver fibrosis, and helped to alleviate the symptoms of bile duct disease [556]. Channel deficiency also shortens the time of hypersensitivity reactions in migraine mouse models by promoting the recovery of normal sensitivity [12]. A cyclic peptide DeC‐1.2 was designed to inhibit ligand activation of TRPM8 but not cold activation, which can eliminate the side effects of cold dysalgesia in oxaliplatin-treated mice without changing body temperature [9]. Analysis of clinical data shows that TRPM8-specific blockers WS12 can reduce tumor growth in colorectal cancer xenografted mice by reducing transcription and activation of Wnt signaling regulators and β-catenin and its target oncogenes, such as C-Myc and Cyclin D1 [732]. TRPML (mucolipin) familyThe TRPML family [782, 1132, 775, 1084, 190] consists of three mammalian members (TRPML1-3). TRPML channels are probably restricted to intracellular vesicles and mutations in the gene (MCOLN1) encoding TRPML1 (mucolipin-1) cause the neurodegenerative disorder mucolipidosis type IV (MLIV) in man. TRPML1 is a cation selective ion channel that is important for sorting/transport of endosomes in the late endocytotic pathway and specifically, fission from late endosome-lysosome hybrid vesicles and lysosomal exocytosis [822]. TRPML2 and TRPML3 show increased channel activity in low luminal sodium and/or increased luminal pH, and are activated by similar small molecules [319, 147, 877]. A naturally occurring gain of function mutation in TRPML3 (i.e. A419P) results in the varitint waddler (Va) mouse phenotype (reviewed by [782, 690]). TRPP (polycystin) familyThe TRPP family (reviewed by [216, 214, 300, 1061, 374]) or PKD2 family is comprised of PKD2 (PC2), PKD2L1 (PC2L1), PKD2L2 (PC2L2), which have been renamed TRPP1, TRPP2 and TRPP3, respectively [1072]. It should also be noted that the nomenclature of PC2 was TRPP2 in old literature. However, PC2 has been uniformed to be called TRPP2 [345]. PKD2 family channels are clearly distinct from the PKD1 family, whose function is unknown. PKD1 and PKD2 form a hetero-oligomeric complex with a 1:3 ratio. [905]. Although still being sorted out, TRPP family members appear to be 6TM spanning nonselective cation channels. TRPV (vanilloid) familyMembers of the TRPV family (reviewed by [995]) can broadly be divided into the non-selective cation channels, TRPV1-4 and the more calcium selective channels TRPV5 and TRPV6. TRPV1-V4 subfamilyTRPV1 is involved in the development of thermal hyperalgesia following inflammation and may contribute to the detection of noxius heat (reviewed by [762, 882, 922]). Numerous splice variants of TRPV1 have been described, some of which modulate the activity of TRPV1, or act in a dominant negative manner when co-expressed with TRPV1 [844]. The pharmacology of TRPV1 channels is discussed in detail in [329] and [1015]. TRPV2 is probably not a thermosensor in man [736], but has recently been implicated in innate immunity [547]. Functional TRPV2 expression is described in placental trophoblast cells of mouse [204]. TRPV3 and TRPV4 are both thermosensitive. There are claims that TRPV4 is also mechanosensitive, but this has not been established to be within a physiological range in a native environment [127, 530]. TRPV5/V6 subfamily TRPV5 and TRPV6 are highly expressed in placenta, bone, and kidney. Under physiological conditions, TRPV5 and TRPV6 are calcium selective channels involved in the absorption and reabsorption of calcium across intestinal and kidney tubule epithelia (reviewed by [1057, 205, 651, 270]).TRPV6 is reported to play a key role in calcium transport in the mouse placenta [1056].
- Published
- 2023
13. LRRC8A anion channels modulate vasodilation via association with Myosin Phosphatase Rho Interacting Protein (MPRIP)
- Author
-
Hyehun Choi, Michael R. Miller, Hong-Ngan Nguyen, Jeffrey C. Rohrbough, Stephen R. Koch, Naoko Boatwright, Michael T. Yarboro, Rajan Sah, W. Hayes McDonald, J. Jeffrey Reese, Ryan J. Stark, and Fred S. Lamb
- Subjects
Article - Abstract
BackgroundIn vascular smooth muscle cells (VSMCs), LRRC8A volume regulated anion channels (VRACs) are activated by inflammatory and pro-contractile stimuli including tumor necrosis factor alpha (TNFα), angiotensin II and stretch. LRRC8A physically associates with NADPH oxidase 1 (Nox1) and supports its production of extracellular superoxide (O2-•).Methods and ResultsMice lacking LRRC8A exclusively in VSMCs (Sm22α-Cre, KO) were used to assess the role of VRACs in TNFα signaling and vasomotor function. KO mesenteric vessels contracted normally to KCl and phenylephrine, but relaxation to acetylcholine (ACh) and sodium nitroprusside (SNP) was enhanced compared to wild type (WT). 48 hours ofex vivoexposure to TNFα (10ng/ml) markedly impaired dilation to ACh and SNP in WT but not KO vessels. VRAC blockade (carbenoxolone, CBX, 100 μM, 20 min) enhanced dilation of control rings and restored impaired dilation following TNFα exposure. Myogenic tone was absent in KO rings. LRRC8A immunoprecipitation followed by mass spectroscopy identified 35 proteins that interacted with LRRC8A. Pathway analysis revealed actin cytoskeletal regulation as the most closely associated function of these proteins. Among these proteins, the Myosin Phosphatase Rho-Interacting protein (MPRIP) links RhoA, MYPT1 and actin. LRRC8A-MPRIP co-localization was confirmed by confocal imaging of tagged proteins, Proximity Ligation Assays, and IP/western blots which revealed LRRC8A binding at the second Pleckstrin Homology domain of MPRIP. siLRRC8A or CBX treatment decreased RhoA activity in cultured VSMCs, and MYPT1 phosphorylation at T853 was reduced in KO mesenteries suggesting that reduced ROCK activity contributes to enhanced relaxation. MPRIP was a target of redox modification, becoming oxidized (sulfenylated) after TNFα exposure.ConclusionsInteraction of Nox1/LRRC8A with MPRIP/RhoA/MYPT1/actin may allow redox regulation of the cytoskeleton and link Nox1 activation to both inflammation and vascular contractility.
- Published
- 2023
14. NON-GENETIC FACTORS AFFECTING GROWTH PERFORMANCE OF INDIGENOUS CHICKEN BREEDS IN NEPAL
- Author
-
Rajan Sah and Roshan Kumar Yadav
- Subjects
business.industry ,Biology ,business ,Indigenous ,Biotechnology - Abstract
There are three identified indigenous poultry breeds in Nepal; Sakini, Ghanti Khuile, Pwankh Ulte. They are reared for dual purpose i.e. meat & egg. To access various non-genetic factors affecting growth performance of indigenous chicken, different relevant research papers, review papers, annual progress reports, and statistical year book were reviewed & analyzed. Results revealed that local breeds are hardy & subject to be affected with environmental factors. They are reared on natural food than formulated feed which meant that they were hardy & rich in high quality protein. They had better weight as they found food continuously in free ranging system. It was found that chicken reared in cool climate had higher body weight than the ones grown in hot period due to less respiratory loss & low body maintenance required. Natural daylight supplemented light at night time was being deployed by farmers & had satisfactory performance. Due to well-developed thick feathers on their body, indigenous chicken were deployed for brooding than artificial methods. Body weight and feed intake was found higher when floor space of 0.279/m2 per bird and flock size of 30 chickens were maintained. Despite any vaccinations, the indigenous breeds still had minimum disease incidence which may be linked to their better adaptability.
- Published
- 2020
15. Smooth Muscle LRRC8A Anion Channel Knockout Promotes Vasodilation and Protects Against TNFα‐induced Vascular Dysfunction
- Author
-
Hyehun Choi, Hong N Nguyen, Rajan Sah, and Fred S. Lamb
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
16. Transient Receptor Potential channels (TRP) in GtoPdb v.2022.1
- Author
-
Michael X. Zhu, Xiaoli Zhang, Lixia Yue, Haoxing Xu, Long-Jun Wu, Joris Vriens, Charlotte Van den Eynde, Dan Tong, Jinbin Tian, Stephanie C. Stotz, Rajan Sah, Antonio Riccio, Grzegorz Owsianik, Elena Oancea, Bernd Nilius, David McKemy, Boyi Liu, Kristopher T Kahle, David Julius, Sven E. Jordt, Kotdaji Ha, Christian M. Grimm, Lu Fan, Julia F. Doerner, Markus Delling, Paul DeCaen, David E. Clapham, Dipayan Chaudhuri, Ingrid Carvacho, and Nathaniel T. Blair
- Abstract
The TRP superfamily of channels (nomenclature as agreed by NC-IUPHAR [159, 999]), whose founder member is the Drosophila Trp channel, exists in mammals as six families; TRPC, TRPM, TRPV, TRPA, TRPP and TRPML based on amino acid homologies. TRP subunits contain six putative TM domains and assemble as homo- or hetero-tetramers to form cation selective channels with diverse modes of activation and varied permeation properties (reviewed by [679]). Established, or potential, physiological functions of the individual members of the TRP families are discussed in detail in the recommended reviews and in a number of books [371, 635, 1066, 236]. The established, or potential, involvement of TRP channels in disease is reviewed in [412, 634] and [637], together with a special edition of Biochemica et Biophysica Acta on the subject [634]. Additional disease related reviews, for pain [585], stroke [1052], sensation and inflammation [921], itch [117], and airway disease [284, 979], are available. The pharmacology of most TRP channels has been advanced in recent years. Broad spectrum agents are listed in the tables along with more selective, or recently recognised, ligands that are flagged by the inclusion of a primary reference. See Rubaiy (2019) for a review of pharmacological tools for TRPC1/C4/C5 channels [751]. Most TRP channels are regulated by phosphoinostides such as PtIns(4,5)P2 although the effects reported are often complex, occasionally contradictory, and likely to be dependent upon experimental conditions, such as intracellular ATP levels (reviewed by [941, 638, 747]). Such regulation is generally not included in the tables.When thermosensitivity is mentioned, it refers specifically to a high Q10 of gating, often in the range of 10-30, but does not necessarily imply that the channel's function is to act as a 'hot' or 'cold' sensor. In general, the search for TRP activators has led to many claims for temperature sensing, mechanosensation, and lipid sensing. All proteins are of course sensitive to energies of binding, mechanical force, and temperature, but the issue is whether the proposed input is within a physiologically relevant range resulting in a response. TRPA (ankyrin) familyTRPA1 is the sole mammalian member of this group (reviewed by [268]). TRPA1 activation of sensory neurons contribute to nociception [382, 831, 555]. Pungent chemicals such as mustard oil (AITC), allicin, and cinnamaldehyde activate TRPA1 by modification of free thiol groups of cysteine side chains, especially those located in its amino terminus [529, 51, 336, 531]. Alkenals with α, β-unsaturated bonds, such as propenal (acrolein), butenal (crotylaldehyde), and 2-pentenal can react with free thiols via Michael addition and can activate TRPA1. However, potency appears to weaken as carbon chain length increases [23, 51]. Covalent modification leads to sustained activation of TRPA1. Chemicals including carvacrol, menthol, and local anesthetics reversibly activate TRPA1 by non-covalent binding [391, 470, 1007, 1006]. TRPA1 is not mechanosensitive under physiological conditions, but can be activated by cold temperatures [392, 193]. The electron cryo-EM structure of TRPA1 [688] indicates that it is a 6-TM homotetramer. Each subunit of the channel contains two short ‘pore helices’ pointing into the ion selectivity filter, which is big enough to allow permeation of partially hydrated Ca2+ ions. TRPC (canonical) familyMembers of the TRPC subfamily (reviewed by [261, 726, 15, 4, 84, 410, 687, 60]) fall into the subgroups outlined below. TRPC2 is a pseudogene in humans. It is generally accepted that all TRPC channels are activated downstream of Gq/11-coupled receptors, or receptor tyrosine kinases (reviewed by [713, 889, 999]). A comprehensive listing of G-protein coupled receptors that activate TRPC channels is given in [4]. Hetero-oligomeric complexes of TRPC channels and their association with proteins to form signalling complexes are detailed in [15] and [411]. TRPC channels have frequently been proposed to act as store-operated channels (SOCs) (or compenents of mulimeric complexes that form SOCs), activated by depletion of intracellular calcium stores (reviewed by [689, 15, 718, 765, 1039, 141, 675, 55, 142]). However, the weight of the evidence is that they are not directly gated by conventional store-operated mechanisms, as established for Stim-gated Orai channels. TRPC channels are not mechanically gated in physiologically relevant ranges of force. All members of the TRPC family are blocked by 2-APB and SKF96365 [319, 318]. Activation of TRPC channels by lipids is discussed by [60]. Important progress has been recently made in TRPC pharmacology [751, 571, 400, 92]. TRPC channels regulate a variety of physiological functions and are implicated in many human diseases [270, 61, 827, 960]. TRPC1/C4/C5 subgroup TRPC1 alone may not form a functional ion channel [210]. TRPC4/C5 may be distinguished from other TRP channels by their potentiation by micromolar concentrations of La3+. TRPC2 is a pseudogene in humans, but in other mammals appears to be an ion channel localized to microvilli of the vomeronasal organ. It is required for normal sexual behavior in response to pheromones in mice. It may also function in the main olfactory epithelia in mice [1036, 672, 673, 1037, 496, 1077, 1032].TRPC3/C6/C7 subgroup All members are activated by diacylglycerol independent of protein kinase C stimulation [319].TRPM (melastatin) familyMembers of the TRPM subfamily (reviewed by [252, 318, 689, 1064]) fall into the five subgroups outlined below. TRPM1/M3 subgroupIn darkness, glutamate released by the photoreceptors and ON-bipolar cells binds to the metabotropic glutamate receptor 6 , leading to activation of Go . This results in the closure of TRPM1. When the photoreceptors are stimulated by light, glutamate release is reduced, and TRPM1 channels are more active, resulting in cell membrane depolarization. Human TRPM1 mutations are associated with congenital stationary night blindness (CSNB), whose patients lack rod function. TRPM1 is also found melanocytes. Isoforms of TRPM1 may present in melanocytes, melanoma, brain, and retina. In melanoma cells, TRPM1 is prevalent in highly dynamic intracellular vesicular structures [368, 657]. TRPM3 (reviewed by [663]) exists as multiple splice variants which differ significantly in their biophysical properties. TRPM3 is expressed in somatosensory neurons and may be important in development of heat hyperalgesia during inflammation (see review [878]). TRPM3 is frequently coexpressed with TRPA1 and TRPV1 in these neurons. TRPM3 is expressed in pancreatic beta cells as well as brain, pituitary gland, eye, kidney, and adipose tissue [662, 877]. TRPM3 may contribute to the detection of noxious heat [949].TRPM2TRPM2 is activated under conditions of oxidative stress (respiratory burst of phagocytic cells) and ischemic conditions. However, the direct activators are ADPR(P) and calcium. As for many ion channels, PIP2 must also be present (reviewed by [1020]). Numerous splice variants of TRPM2 exist which differ in their activation mechanisms [219]. The C-terminal domain contains a TRP motif, a coiled-coil region, and an enzymatic NUDT9 homologous domain. TRPM2 appears not to be activated by NAD, NAAD, or NAADP, but is directly activated by ADPRP (adenosine-5'-O-disphosphoribose phosphate) [902]. TRPM2 is involved in warmth sensation [789], and contributes to neurological diseases [66]. Recent study shows that 2'-deoxy-ADPR is an endogenous TRPM2 superagonist [253]. TRPM4/5 subgroupTRPM4 and TRPM5 have the distinction within all TRP channels of being impermeable to Ca2+ [999]. A splice variant of TRPM4 (i.e.TRPM4b) and TRPM5 are molecular candidates for endogenous calcium-activated cation (CAN) channels [301]. TRPM4 is active in the late phase of repolarization of the cardiac ventricular action potential. TRPM4 deletion or knockout enhances beta adrenergic-mediated inotropy [546]. Mutations are associated with conduction defects [374, 546, 821]. TRPM4 has been shown to be an important regulator of Ca2+ entry in to mast cells [926] and dendritic cell migration [43]. TRPM5 in taste receptor cells of the tongue appears essential for the transduction of sweet, amino acid and bitter stimuli [494] TRPM5 contributes to the slow afterdepolarization of layer 5 neurons in mouse prefrontal cortex [471]. Both TRPM4 and TRPM5 are required transduction of taste stimuli [226].TRPM6/7 subgroupTRPM6 and 7 combine channel and enzymatic activities (‘chanzymes’). These channels have the unusual property of permeation by divalent (Ca2+, Mg2+, Zn2+) and monovalent cations, high single channel conductances, but overall extremely small inward conductance when expressed to the plasma membrane. They are inhibited by internal Mg2+ at ~0.6 mM, around the free level of Mg2+ in cells. Whether they contribute to Mg2+ homeostasis is a contentious issue. When either gene is deleted in mice, the result is embryonic lethality. The C-terminal kinase region is cleaved under unknown stimuli, and the kinase phosphorylates nuclear histones. TRPM7 is responsible for oxidant- induced Zn2+ release from intracellular vesicles [3] and contributes to intestinal mineral absorption essential for postnatal survival [574]. TRPM8Is a channel activated by cooling and pharmacological agents evoking a ‘cool’ sensation and participates in the thermosensation of cold temperatures [54, 161, 205] reviewed by [943, 516, 420, 599]. TRPML (mucolipin) familyThe TRPML family [729, 1049, 723, 1010, 173] consists of three mammalian members (TRPML1-3). TRPML channels are probably restricted to intracellular vesicles and mutations in the gene (MCOLN1) encoding TRPML1 (mucolipin-1) cause the neurodegenerative disorder mucolipidosis type IV (MLIV) in man. TRPML1 is a cation selective ion channel that is important for sorting/transport of endosomes in the late endocytotic pathway and specifically, fission from late endosome-lysosome hybrid vesicles and lysosomal exocytosis [766]. TRPML2 and TRPML3 show increased channel activity in low extracellular sodium and are activated by similar small molecules [293]. A naturally occurring gain of function mutation in TRPML3 (i.e. A419P) results in the varitint waddler (Va) mouse phenotype (reviewed by [729, 639]). TRPP (polycystin) familyThe TRPP family (reviewed by [197, 195, 275, 988, 345]) or PKD2 family is comprised of PKD2 (PC2), PKD2L1 (PC2L1), PKD2L2 (PC2L2), which have been renamed TRPP1, TRPP2 and TRPP3, respectively [999]. It should also be noted that the nomenclature of PC2 was TRPP2 in old literature. However, PC2 has been uniformed to be called TRPP2 [317]. PKD2 family channels are clearly distinct from the PKD1 family, whose function is unknown. PKD1 and PKD2 form a hetero-oligomeric complex with a 1:3 ratio. [845]. Although still being sorted out, TRPP family members appear to be 6TM spanning nonselective cation channels. TRPV (vanilloid) familyMembers of the TRPV family (reviewed by [928]) can broadly be divided into the non-selective cation channels, TRPV1-4 and the more calcium selective channels TRPV5 and TRPV6.TRPV1-V4 subfamilyTRPV1 is involved in the development of thermal hyperalgesia following inflammation and may contribute to the detection of noxius heat (reviewed by [710, 824, 860]). Numerous splice variants of TRPV1 have been described, some of which modulate the activity of TRPV1, or act in a dominant negative manner when co-expressed with TRPV1 [787]. The pharmacology of TRPV1 channels is discussed in detail in [303] and [947]. TRPV2 is probably not a thermosensor in man [684], but has recently been implicated in innate immunity [503]. TRPV3 and TRPV4 are both thermosensitive. There are claims that TRPV4 is also mechanosensitive, but this has not been established to be within a physiological range in a native environment [114, 488].TRPV5/V6 subfamily TRPV5 and TRPV6 are highly expressed in placenta, bone, and kidney. Under physiological conditions, TRPV5 and TRPV6 are calcium selective channels involved in the absorption and reabsorption of calcium across intestinal and kidney tubule epithelia (reviewed by [984, 185, 601, 248]).
- Published
- 2022
17. LRRC8 family proteins within lysosomes regulate cellular osmoregulation and enhance cell survival to multiple physiological stresses
- Author
-
Timothy L. Cover, Zhuang Zhuang Zhao, Mingxue Gu, Raif S. Geha, Rajan Sah, Janet Chou, Richard I. Hume, Haoxing Xu, Yexin Yang, Ping Li, Nirakar Sahoo, Fernando Benavides, Shiyu Xiao, Xinghua Feng, Meiqin Hu, Ce Wang, and Ying Yang
- Subjects
Anions ,Cell Survival ,Vacuole ,Exocytosis ,Gene Knockout Techniques ,Mice ,Osmoregulation ,Stress, Physiological ,Lysosome ,Chlorocebus aethiops ,medicine ,Animals ,Homeostasis ,Humans ,Multidisciplinary ,Chemistry ,Membrane Proteins ,Biological Sciences ,Cell biology ,Cytosol ,HEK293 Cells ,medicine.anatomical_structure ,Cytoplasm ,COS Cells ,Vacuoles ,lipids (amino acids, peptides, and proteins) ,Lysosomes ,Transcriptome ,Intracellular - Abstract
LRRC8 family proteins on the plasma membrane play a critical role in cellular osmoregulation by forming volume-regulated anion channels (VRACs) necessary to prevent necrotic cell death. We demonstrate that intracellular LRRC8 proteins acting within lysosomes also play an essential role in cellular osmoregulation. LRRC8 proteins on lysosome membranes generate large lysosomal volume-regulated anion channel (Lyso-VRAC) currents in response to low cytoplasmic ionic strength conditions. When a double-leucine L(706)L(707) motif at the C terminus of LRRC8A was mutated to alanines, normal plasma membrane VRAC currents were still observed, but Lyso-VRAC currents were absent. We used this targeting mutant, as well as pharmacological tools, to demonstrate that Lyso-VRAC currents are necessary for the formation of large lysosome-derived vacuoles, which store and then expel excess water to maintain cytosolic water homeostasis. Thus, Lyso-VRACs allow lysosomes of mammalian cells to act as the cell`s “bladder.” When Lyso-VRAC current was selectively eliminated, the extent of necrotic cell death to sustained stress was greatly increased, not only in response to hypoosmotic stress, but also to hypoxic and hypothermic stresses. Thus Lyso-VRACs play an essential role in enabling cells to mount successful homeostatic responses to multiple stressors.
- Published
- 2020
18. Links between ceramides and cardiac function
- Author
-
Lauren K. Park, Valene Garr Barry, Juan Hong, John Heebink, Rajan Sah, and Linda R. Peterson
- Subjects
Heart Failure ,Nutrition and Dietetics ,Endocrinology, Diabetes and Metabolism ,Heart ,Cell Biology ,Ceramides ,Article ,Ventricular Dysfunction, Left ,Genetics ,Animals ,Humans ,Cardiology and Cardiovascular Medicine ,Molecular Biology ,Biomarkers - Abstract
Total ceramide levels in cardiac tissue relate to cardiac dysfunction in animal models. However, emerging evidence suggests that the fatty acyl chain length of ceramides also impacts their relationship to cardiac function. This review explores evidence regarding the relationship between ceramides and left ventricular dysfunction and heart failure. It further explores possible mechanisms underlying these relationships.In large, community-based cohorts, a higher ratio of specific plasma ceramides, C16 : 0/C24 : 0, related to worse left ventricular dysfunction. Increased left ventricular mass correlated with plasma C16 : 0/C24 : 0, but this relationship became nonsignificant after adjustment for multiple comparisons. Decreased left atrial function and increased left atrial size also related to C16 : 0/C24 : 0. Furthermore, increased incident heart failure, overall cardiovascular disease (CVD) mortality and all-cause mortality were associated with higher C16 : 0/C24 : 0 (or lower C24 : 0/C16 : 0). Finally, a number of possible biological mechanisms are outlined supporting the link between C16 : 0/C24 : 0 ceramides, ceramide signalling and CVD.High cardiac levels of total ceramides are noted in heart failure. In the plasma, C16 : 0/C24 : 0 ceramides may be a valuable biomarker of preclinical left ventricular dysfunction, remodelling, heart failure and mortality. Continued exploration of the mechanisms underlying these profound relationships may help develop specific lipid modulators to combat cardiac dysfunction and heart failure.
- Published
- 2022
19. Transient Receptor Potential channels (TRP) in GtoPdb v.2021.3
- Author
-
Sven E. Jordt, Antonio Riccio, Elena Oancea, David Julius, Haoxing Xu, Ingrid Carvacho, Rajan Sah, Julia F. Doerner, Kotdaji Ha, Boyi Liu, Dan Tong, Charlotte Van den Eynde, David D. McKemy, Jinbin Tian, Paul G. DeCaen, Stephanie C. Stotz, Dipayan Chaudhuri, Grzegorz Owsianik, Joris Vriens, Long Jun Wu, Michael X. Zhu, Lu Fan, Bernd Nilius, Markus Delling, Kristopher T. Kahle, Lixia Yue, Nathaniel T. Blair, Xiaoli Zhang, and David E. Clapham
- Subjects
Transient receptor potential channel ,Chemistry ,Biophysics - Abstract
The TRP superfamily of channels (nomenclature as agreed by NC-IUPHAR [159, 997]), whose founder member is the Drosophila Trp channel, exists in mammals as six families; TRPC, TRPM, TRPV, TRPA, TRPP and TRPML based on amino acid homologies. TRP subunits contain six putative TM domains and assemble as homo- or hetero-tetramers to form cation selective channels with diverse modes of activation and varied permeation properties (reviewed by [679]). Established, or potential, physiological functions of the individual members of the TRP families are discussed in detail in the recommended reviews and in a number of books [371, 635, 1064, 236]. The established, or potential, involvement of TRP channels in disease is reviewed in [412, 634] and [637], together with a special edition of Biochemica et Biophysica Acta on the subject [634]. Additional disease related reviews, for pain [585], stroke [1050], sensation and inflammation [919], itch [117], and airway disease [284, 977], are available. The pharmacology of most TRP channels has been advanced in recent years. Broad spectrum agents are listed in the tables along with more selective, or recently recognised, ligands that are flagged by the inclusion of a primary reference. See Rubaiy (2019) for a review of pharmacological tools for TRPC1/C4/C5 channels [751]. Most TRP channels are regulated by phosphoinostides such as PtIns(4,5)P2 although the effects reported are often complex, occasionally contradictory, and likely to be dependent upon experimental conditions, such as intracellular ATP levels (reviewed by [939, 638, 747]). Such regulation is generally not included in the tables.When thermosensitivity is mentioned, it refers specifically to a high Q10 of gating, often in the range of 10-30, but does not necessarily imply that the channel's function is to act as a 'hot' or 'cold' sensor. In general, the search for TRP activators has led to many claims for temperature sensing, mechanosensation, and lipid sensing. All proteins are of course sensitive to energies of binding, mechanical force, and temperature, but the issue is whether the proposed input is within a physiologically relevant range resulting in a response. TRPA (ankyrin) familyTRPA1 is the sole mammalian member of this group (reviewed by [268]). TRPA1 activation of sensory neurons contribute to nociception [382, 829, 555]. Pungent chemicals such as mustard oil (AITC), allicin, and cinnamaldehyde activate TRPA1 by modification of free thiol groups of cysteine side chains, especially those located in its amino terminus [529, 51, 336, 531]. Alkenals with α, β-unsaturated bonds, such as propenal (acrolein), butenal (crotylaldehyde), and 2-pentenal can react with free thiols via Michael addition and can activate TRPA1. However, potency appears to weaken as carbon chain length increases [23, 51]. Covalent modification leads to sustained activation of TRPA1. Chemicals including carvacrol, menthol, and local anesthetics reversibly activate TRPA1 by non-covalent binding [391, 470, 1005, 1004]. TRPA1 is not mechanosensitive under physiological conditions, but can be activated by cold temperatures [392, 193]. The electron cryo-EM structure of TRPA1 [688] indicates that it is a 6-TM homotetramer. Each subunit of the channel contains two short ‘pore helices’ pointing into the ion selectivity filter, which is big enough to allow permeation of partially hydrated Ca2+ ions. TRPC (canonical) familyMembers of the TRPC subfamily (reviewed by [261, 726, 15, 4, 84, 410, 687, 60]) fall into the subgroups outlined below. TRPC2 is a pseudogene in humans. It is generally accepted that all TRPC channels are activated downstream of Gq/11-coupled receptors, or receptor tyrosine kinases (reviewed by [713, 887, 997]). A comprehensive listing of G-protein coupled receptors that activate TRPC channels is given in [4]. Hetero-oligomeric complexes of TRPC channels and their association with proteins to form signalling complexes are detailed in [15] and [411]. TRPC channels have frequently been proposed to act as store-operated channels (SOCs) (or compenents of mulimeric complexes that form SOCs), activated by depletion of intracellular calcium stores (reviewed by [689, 15, 718, 764, 1037, 141, 675, 55, 142]). However, the weight of the evidence is that they are not directly gated by conventional store-operated mechanisms, as established for Stim-gated Orai channels. TRPC channels are not mechanically gated in physiologically relevant ranges of force. All members of the TRPC family are blocked by 2-APB and SKF96365 [319, 318]. Activation of TRPC channels by lipids is discussed by [60]. Important progress has been recently made in TRPC pharmacology [751, 571, 400, 92]. TRPC channels regulate a variety of physiological functions and are implicated in many human diseases [270, 61, 825, 958]. TRPC1/C4/C5 subgroup TRPC1 alone may not form a functional ion channel [210]. TRPC4/C5 may be distinguished from other TRP channels by their potentiation by micromolar concentrations of La3+. TRPC2 is a pseudogene in humans, but in other mammals appears to be an ion channel localized to microvilli of the vomeronasal organ. It is required for normal sexual behavior in response to pheromones in mice. It may also function in the main olfactory epithelia in mice [1034, 672, 673, 1035, 496, 1075, 1030].TRPC3/C6/C7 subgroup All members are activated by diacylglycerol independent of protein kinase C stimulation [319].TRPM (melastatin) familyMembers of the TRPM subfamily (reviewed by [252, 318, 689, 1062]) fall into the five subgroups outlined below. TRPM1/M3 subgroupIn darkness, glutamate released by the photoreceptors and ON-bipolar cells binds to the metabotropic glutamate receptor 6 , leading to activation of Go . This results in the closure of TRPM1. When the photoreceptors are stimulated by light, glutamate release is reduced, and TRPM1 channels are more active, resulting in cell membrane depolarization. Human TRPM1 mutations are associated with congenital stationary night blindness (CSNB), whose patients lack rod function. TRPM1 is also found melanocytes. Isoforms of TRPM1 may present in melanocytes, melanoma, brain, and retina. In melanoma cells, TRPM1 is prevalent in highly dynamic intracellular vesicular structures [368, 657]. TRPM3 (reviewed by [663]) exists as multiple splice variants which differ significantly in their biophysical properties. TRPM3 is expressed in somatosensory neurons and may be important in development of heat hyperalgesia during inflammation (see review [876]). TRPM3 is frequently coexpressed with TRPA1 and TRPV1 in these neurons. TRPM3 is expressed in pancreatic beta cells as well as brain, pituitary gland, eye, kidney, and adipose tissue [662, 875]. TRPM3 may contribute to the detection of noxious heat [947].TRPM2TRPM2 is activated under conditions of oxidative stress (respiratory burst of phagocytic cells) and ischemic conditions. However, the direct activators are ADPR(P) and calcium. As for many ion channels, PIP2 must also be present (reviewed by [1018]). Numerous splice variants of TRPM2 exist which differ in their activation mechanisms [219]. The C-terminal domain contains a TRP motif, a coiled-coil region, and an enzymatic NUDT9 homologous domain. TRPM2 appears not to be activated by NAD, NAAD, or NAADP, but is directly activated by ADPRP (adenosine-5'-O-disphosphoribose phosphate) [900]. TRPM2 is involved in warmth sensation [788], and contributes to neurological diseases [66]. Recent study shows that 2'-deoxy-ADPR is an endogenous TRPM2 superagonist [253]. TRPM4/5 subgroupTRPM4 and TRPM5 have the distinction within all TRP channels of being impermeable to Ca2+ [997]. A splice variant of TRPM4 (i.e.TRPM4b) and TRPM5 are molecular candidates for endogenous calcium-activated cation (CAN) channels [301]. TRPM4 is active in the late phase of repolarization of the cardiac ventricular action potential. TRPM4 deletion or knockout enhances beta adrenergic-mediated inotropy [546]. Mutations are associated with conduction defects [374, 546, 819]. TRPM4 has been shown to be an important regulator of Ca2+ entry in to mast cells [924] and dendritic cell migration [43]. TRPM5 in taste receptor cells of the tongue appears essential for the transduction of sweet, amino acid and bitter stimuli [494] TRPM5 contributes to the slow afterdepolarization of layer 5 neurons in mouse prefrontal cortex [471]. Both TRPM4 and TRPM5 are required transduction of taste stimuli [226].TRPM6/7 subgroupTRPM6 and 7 combine channel and enzymatic activities (‘chanzymes’). These channels have the unusual property of permeation by divalent (Ca2+, Mg2+, Zn2+) and monovalent cations, high single channel conductances, but overall extremely small inward conductance when expressed to the plasma membrane. They are inhibited by internal Mg2+ at ~0.6 mM, around the free level of Mg2+ in cells. Whether they contribute to Mg2+ homeostasis is a contentious issue. When either gene is deleted in mice, the result is embryonic lethality. The C-terminal kinase region is cleaved under unknown stimuli, and the kinase phosphorylates nuclear histones. TRPM7 is responsible for oxidant- induced Zn2+ release from intracellular vesicles [3] and contributes to intestinal mineral absorption essential for postnatal survival [574]. TRPM8Is a channel activated by cooling and pharmacological agents evoking a ‘cool’ sensation and participates in the thermosensation of cold temperatures [54, 161, 205] reviewed by [941, 516, 420, 599]. TRPML (mucolipin) familyThe TRPML family [729, 1047, 723, 1008, 173] consists of three mammalian members (TRPML1-3). TRPML channels are probably restricted to intracellular vesicles and mutations in the gene (MCOLN1) encoding TRPML1 (mucolipin-1) cause the neurodegenerative disorder mucolipidosis type IV (MLIV) in man. TRPML1 is a cation selective ion channel that is important for sorting/transport of endosomes in the late endocytotic pathway and specifically, fission from late endosome-lysosome hybrid vesicles and lysosomal exocytosis [765]. TRPML2 and TRPML3 show increased channel activity in low extracellular sodium and are activated by similar small molecules [293]. A naturally occurring gain of function mutation in TRPML3 (i.e. A419P) results in the varitint waddler (Va) mouse phenotype (reviewed by [729, 639]). TRPP (polycystin) familyThe TRPP family (reviewed by [197, 195, 275, 986, 345]) or PKD2 family is comprised of PKD2 (PC2), PKD2L1 (PC2L1), PKD2L2 (PC2L2), which have been renamed TRPP1, TRPP2 and TRPP3, respectively [997]. It should also be noted that the nomenclature of PC2 was TRPP2 in old literature. However, PC2 has been uniformed to be called TRPP2 [317]. PKD2 family channels are clearly distinct from the PKD1 family, whose function is unknown. PKD1 and PKD2 form a hetero-oligomeric complex with a 1:3 ratio. [843]. Although still being sorted out, TRPP family members appear to be 6TM spanning nonselective cation channels. TRPV (vanilloid) familyMembers of the TRPV family (reviewed by [926]) can broadly be divided into the non-selective cation channels, TRPV1-4 and the more calcium selective channels TRPV5 and TRPV6.TRPV1-V4 subfamilyTRPV1 is involved in the development of thermal hyperalgesia following inflammation and may contribute to the detection of noxius heat (reviewed by [710, 822, 858]). Numerous splice variants of TRPV1 have been described, some of which modulate the activity of TRPV1, or act in a dominant negative manner when co-expressed with TRPV1 [786]. The pharmacology of TRPV1 channels is discussed in detail in [303] and [945]. TRPV2 is probably not a thermosensor in man [684], but has recently been implicated in innate immunity [503]. TRPV3 and TRPV4 are both thermosensitive. There are claims that TRPV4 is also mechanosensitive, but this has not been established to be within a physiological range in a native environment [114, 488].TRPV5/V6 subfamily TRPV5 and TRPV6 are highly expressed in placenta, bone, and kidney. Under physiological conditions, TRPV5 and TRPV6 are calcium selective channels involved in the absorption and reabsorption of calcium across intestinal and kidney tubule epithelia (reviewed by [982, 185, 601, 248]).
- Published
- 2021
20. Electrophysiological and Molecular Mechanisms of Sinoatrial Node Mechanosensitivity
- Author
-
Chen Kang, Alexey V. Glukhov, Daniel Turner, Matteo E. Mangoni, Juan Hong, Rajan Sah, P. Mesirca, University of Wisconsin School of Medicine and Public Health, Washington University School of Medicine in St. Louis, Washington University in Saint Louis (WUSTL), Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), Mangoni, Matteo, and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
cardiac ,Cardiovascular Medicine ,030204 cardiovascular system & hematology ,automaticity ,03 medical and health sciences ,Transient receptor potential channel ,0302 clinical medicine ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Hypothesis and Theory ,Caveolae ,heart rate ,medicine ,Diseases of the circulatory (Cardiovascular) system ,Ion channel ,030304 developmental biology ,0303 health sciences ,calcium ,Chemistry ,Sinoatrial node ,stretch activated ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Electrophysiology ,medicine.anatomical_structure ,RC666-701 ,ion channel ,Chloride channel ,Mechanosensitive channels ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Neuroscience - Abstract
International audience; The understanding of the electrophysiological mechanisms that underlie mechanosensitivity of the sinoatrial node (SAN), the primary pacemaker of the heart, has been evolving over the past century. The heart is constantly exposed to a dynamic mechanical environment; as such, the SAN has numerous canonical and emerging mechanosensitive ion channels and signaling pathways that govern its ability to respond to both fast (within second or on beat-to-beat manner) and slow (minutes) timescales. This review summarizes the effects of mechanical loading on the SAN activity and reviews putative candidates, including fast mechanoactivated channels (Piezo, TREK, and BK) and slow mechanoresponsive ion channels [including volume-regulated chloride channels and transient receptor potential (TRP)], as well as the components of mechanochemical signal transduction, which may contribute to SAN mechanosensitivity. Furthermore, we examine the structural foundation for both mechano-electrical and mechanochemical signal transduction and discuss the role of specialized membrane nanodomains, namely, caveolae, in mechanical regulation of both membrane and calcium clock components of the so-called coupled-clock pacemaker system responsible for SAN automaticity. Finally, we emphasize how these mechanically activated changes contribute to the pathophysiology of SAN dysfunction and discuss controversial areas necessitating future investigations. Though the exact mechanisms of SAN mechanosensitivity are currently unknown, identification of such components, their impact into SAN pacemaking, and pathological remodeling may provide new therapeutic targets for the treatment of SAN dysfunction and associated rhythm abnormalities.
- Published
- 2021
21. Late adolescence mortality in mice with brain-specific deletion of the volume-regulated anion channel subunit LRRC8A
- Author
-
Yunfei Huang, Corinne S. Wilson, Julia W. Nalwalk, Alexander A. Mongin, Rajan Sah, Preeti Dohare, Russell J. Ferland, Shaina Orbeta, and Annalisa Scimemi
- Subjects
Male ,Glutamate-glutamine cycle ,Glutamic Acid ,Biochemistry ,Mice ,Slice preparation ,Seizures ,Glutamine synthetase ,Genetics ,medicine ,GABA transporter ,Animals ,Gliosis ,Molecular Biology ,CA1 Region, Hippocampal ,Mice, Knockout ,Ion Transport ,biology ,Chemistry ,Glutamate receptor ,Membrane Proteins ,medicine.disease ,Astrogliosis ,Cell biology ,Mice, Inbred C57BL ,Astrocytes ,Synaptic plasticity ,biology.protein ,GABAergic ,Female ,Biotechnology - Abstract
The leucine-rich repeat-containing family 8 member A (LRRC8A) is an essential subunit of the volume-regulated anion channel (VRAC). VRAC is critical for cell volume control, but its broader physiological functions remain under investigation. Recent studies in the field indicate that Lrrc8a disruption in the brain astrocytes reduces neuronal excitability, impairs synaptic plasticity and memory, and protects against cerebral ischemia. In the present work, we generated brain-wide conditional LRRC8A knockout mice (LRRC8A bKO) using NestinCre -driven Lrrc8aflox/flox excision in neurons, astrocytes, and oligodendroglia. LRRC8A bKO animals were born close to the expected Mendelian ratio and developed without overt histological abnormalities, but, surprisingly, all died between 5 and 9 weeks of age with a seizure phenotype, which was confirmed by video and EEG recordings. Brain slice electrophysiology detected changes in the excitability of pyramidal cells and modified GABAergic inputs in the hippocampal CA1 region of LRRC8A bKO. LRRC8A-null hippocampi showed increased immunoreactivity of the astrocytic marker GFAP, indicating reactive astrogliosis. We also found decreased whole-brain protein levels of the GABA transporter GAT-1, the glutamate transporter GLT-1, and the astrocytic enzyme glutamine synthetase. Complementary HPLC assays identified reduction in the tissue levels of the glutamate and GABA precursor glutamine. Together, these findings suggest that VRAC provides vital control of brain excitability in mouse adolescence. VRAC deletion leads to a lethal phenotype involving progressive astrogliosis and dysregulation of astrocytic uptake and supply of amino acid neurotransmitters and their precursors.
- Published
- 2021
22. SWELL signalling in adipocytes: can fat 'feel' fat?
- Author
-
Litao Xie, Susheel K. Gunasekar, and Rajan Sah
- Subjects
medicine.medical_specialty ,obesity ,insulin ,Histology ,medicine.medical_treatment ,Disease ,Type 2 diabetes ,Osteoarthritis ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,mechano-transduction ,lcsh:Physiology ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Caveolae ,Internal medicine ,Adipocytes ,medicine ,Animals ,Homeostasis ,Humans ,lcsh:QH573-671 ,030304 developmental biology ,0303 health sciences ,lcsh:RC648-665 ,lcsh:QP1-981 ,business.industry ,lcsh:Cytology ,Insulin ,Fatty liver ,Membrane Proteins ,Cancer ,Lipid Droplets ,Cell Biology ,medicine.disease ,Obesity ,3. Good health ,Glucose ,Endocrinology ,ion channel ,caveolae ,Commentary ,business ,Proto-Oncogene Proteins c-akt ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Obesity is becoming a global epidemic, predisposing to Type 2 diabetes, cardiovascular disease, fatty liver disease, pulmonary disease, osteoarthritis and cancer. Therefore, understanding the biology of adipocyte expansion in response to overnutrition is critical to devising strategies to treat obesity, and the associated burden of morbidity and mortality. Through exploratory patch-clamp experiments in freshly isolated primary murine and human adipocytes, we recently determined that SWELL1/LRRC8a, a leucine-rich repeat containing transmembrane protein, functionally encoded an ion channel signalling complex (the volume-regulated anion channel, or VRAC) on the adipocyte plasma membrane. The SWELL1-/LRRC8 channel complex activates in response to increases in adipocyte volume and in the context of obesity. SWELL1 is also required for insulin-PI3K-AKT2 signalling to regulate adipocyte growth and systemic glycaemia. This commentary delves further into our working models for the molecular mechanisms of adipocyte SWELL1-mediated VRAC activation, proposed signal transduction mechanisms, and putative impact on adipocyte hypertrophy during caloric excess.
- Published
- 2019
23. Decision letter: The molecular appearance of native TRPM7 channel complexes identified by high-resolution proteomics
- Author
-
Rajan Sah, Thomas Voets, and László Csanády
- Subjects
Physics ,TRPM7 ,High resolution ,Channel (broadcasting) ,Biological system ,Proteomics - Published
- 2021
24. Perilipin 2 downregulation in β cells impairs insulin secretion under nutritional stress and damages mitochondria
- Author
-
Siming Liu, James A. Ankrum, Brian D. Fink, Akansha Mishra, Laura Jackson, Frederick Anokye-Danso, Gourav Bhardwaj, Yumi Imai, Chen Kang, Samuel B. Stephens, Timothy H. King, Stefan Strack, Mikako Harata, Joseph A. Promes, Andrew S. Greenberg, Rexford S. Ahima, Brian T. O’Neill, Rajan Sah, and William I. Sivitz
- Subjects
0301 basic medicine ,Mitochondrion ,Oxidative Phosphorylation ,Mice ,0302 clinical medicine ,Endocrinology ,Insulin-Secreting Cells ,Lipid droplet ,Insulin Secretion ,Mice, Knockout ,biology ,Chemistry ,Diabetes ,Islet cells ,General Medicine ,Mitochondria ,030220 oncology & carcinogenesis ,Medicine ,Research Article ,medicine.medical_specialty ,endocrine system ,Perilipin 2 ,Down-Regulation ,Oxidative phosphorylation ,In Vitro Techniques ,Diet, High-Fat ,Perilipin-2 ,Islets of Langerhans ,03 medical and health sciences ,Oxygen Consumption ,Downregulation and upregulation ,Stress, Physiological ,Carnitine ,Internal medicine ,medicine ,Animals ,Humans ,Fragmentation (cell biology) ,Lipid Droplets ,Metabolism ,In vitro ,Rats ,Oxidative Stress ,Glucose ,030104 developmental biology ,biology.protein ,Oleic Acid - Abstract
Perilipin 2 (PLIN2) is a lipid droplet (LD) protein in β cells that increases under nutritional stress. Downregulation of PLIN2 is often sufficient to reduce LD accumulation. To determine whether PLIN2 positively or negatively affects β cell function under nutritional stress, PLIN2 was downregulated in mouse β cells, INS1 cells, and human islet cells. β Cell–specific deletion of PLIN2 in mice on a high-fat diet reduced glucose-stimulated insulin secretion (GSIS) in vivo and in vitro. Downregulation of PLIN2 in INS1 cells blunted GSIS after 24-hour incubation with 0.2 mM palmitic acid. Downregulation of PLIN2 in human pseudoislets cultured at 5.6 mM glucose impaired both phases of GSIS, indicating that PLIN2 is critical for GSIS. Downregulation of PLIN2 decreased specific OXPHOS proteins in all 3 models and reduced oxygen consumption rates in INS1 cells and mouse islets. Moreover, we found that PLIN2-deficient INS1 cells increased the distribution of a fluorescent oleic acid analog to mitochondria and showed signs of mitochondrial stress, as indicated by susceptibility to fragmentation and alterations of acyl-carnitines and glucose metabolites. Collectively, PLIN2 in β cells has an important role in preserving insulin secretion, β cell metabolism, and mitochondrial function under nutritional stress.
- Published
- 2021
25. Mechanosensitive TRPV4 is required for crystal-induced inflammation
- Author
-
Ping Lu, Xueping Yue, Jialie Luo, Rajan Sah, Peng Liu, Jing Feng, Zhe Zhu, Xueming Hu, Brian S. Kim, Zhou Lan, Kory J. Lavine, Zhiyong Liu, Pu Yang, Yonghui Zhao, Fang Wang, Lixia Du, Lvyi Chen, Hongzhen Hu, Liang Cao, and Zili Xie
- Subjects
Adult ,Male ,Patch-Clamp Techniques ,Crystal Arthropathies ,Gout ,Inflammasomes ,THP-1 Cells ,medicine.medical_treatment ,Immunology ,Interleukin-1beta ,Arthritis ,TRPV Cation Channels ,Inflammation ,Peripheral blood mononuclear cell ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,Rheumatology ,Downregulation and upregulation ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Immunology and Allergy ,Animals ,Humans ,Innate immune system ,business.industry ,Arthritis, Gouty ,Macrophages ,Optical Imaging ,Synovial Membrane ,Nociceptors ,Inflammasome ,Middle Aged ,medicine.disease ,Arthralgia ,Uric Acid ,Cytokine ,Leukocytes, Mononuclear ,Mechanosensitive channels ,medicine.symptom ,business ,medicine.drug - Abstract
Crystal structures activate innate immune cells, especially macrophages and initiate inflammatory responses. We aimed to understand the role of the mechanosensitive TRPV4 channel in crystal-induced inflammation. Real-time RT-PCR, RNAscope in situ hybridisation, and Trpv4eGFP mice were used to examine TRPV4 expression and whole-cell patch-clamp recording and live-cell Ca2+ imaging were used to study TRPV4 function in mouse synovial macrophages and human peripheral blood mononuclear cells (PBMCs). Both genetic deletion and pharmacological inhibition approaches were used to investigate the role of TRPV4 in NLRP3 inflammasome activation induced by diverse crystals in vitro and in mouse models of crystal-induced pain and inflammation in vivo. TRPV4 was functionally expressed by synovial macrophages and human PBMCs and TRPV4 expression was upregulated by stimulation with monosodium urate (MSU) crystals and in human PBMCs from patients with acute gout flares. MSU crystal-induced gouty arthritis were significantly reduced by either genetic ablation or pharmacological inhibition of TRPV4 function. Mechanistically, TRPV4 mediated the activation of NLRP3 inflammasome by diverse crystalline materials but not non-crystalline NLRP3 inflammasome activators, driving the production of inflammatory cytokine interleukin-1β which elicited TRPV4-dependent inflammatory responses in vivo. Moreover, chemical ablation of the TRPV1-expressing nociceptors significantly attenuated the MSU crystal-induced gouty arthritis. In conclusion, TRPV4 is a common mediator of inflammatory responses induced by diverse crystals through NLRP3 inflammasome activation in macrophages. TRPV4-expressing resident macrophages are critically involved in MSU crystal-induced gouty arthritis. A neuroimmune interaction between the TRPV1-expressing nociceptors and the TRPV4-expressing synovial macrophages contributes to the generation of acute gout flares.
- Published
- 2021
26. Small molecule SWELL1-LRRC8 complex induction improves glycemic control and nonalcoholic fatty liver disease in murine Type 2 diabetes
- Author
-
Yumi Imai, Eric B. Taylor, Stephen G. Brohawn, Susheel K. Gunasekar, Norris Aw, Isaac Samuel, Pratik Rajesh Chheda, Ashok Kumar, David M Kern, Macaulay Elliot-Hudson, Robert J. Kerns, Joshua M Maurer, Rajan Sah, Chen Kang, Jessica K. Smith, Chaitanya A. Kulkarni, Grzesik Wj, My-Ta C, Litao Xie, Peter Nau, Sheldon Rd, E.E. Gerber, Lerner Dj, and Yuyun Zhang
- Subjects
medicine.medical_specialty ,geography ,geography.geographical_feature_category ,Chemistry ,nutritional and metabolic diseases ,Adipose tissue ,Skeletal muscle ,Type 2 diabetes ,medicine.disease ,Islet ,Pathogenesis ,Endocrinology ,medicine.anatomical_structure ,Insulin resistance ,Internal medicine ,Nonalcoholic fatty liver disease ,medicine ,Glycemic - Abstract
Type 2 diabetes (T2D) is associated with insulin resistance, impaired insulin secretion from the pancreatic β-cell, and nonalcoholic fatty liver disease (NAFLD). SWELL1 (LRRC8a) ablation impairs adipose and skeletal muscle insulin-pAKT2 signaling, β-cell insulin secretion and glycemic control - suggesting that SWELL1-LRRC8 complex dysfunction contributes to T2D pathogenesis. Here, we show that ICl,SWELLand SWELL1 protein are reduced in adipose and β-cells in murine and human T2D. Combining cryo-electron microscopy, molecular docking, medicinal chemistry, and functional studies, we define a structure activity relationship to rationally-designed active derivatives (SN-40X) of a SWELL1 channel inhibitor (DCPIB/SN-401), that bind the SWELL1-LRRC8 hexameric complex, restore SWELL1-LRRC8 protein, plasma membrane trafficking, signaling and islet insulin secretion via SWELL1-dependent mechanisms.In vivo, SN-401 and active SN-40X compounds restore glycemic control and prevents NAFLD by improving insulin-sensitivity and insulin secretion in murine T2D. These findings demonstrate that small molecule SWELL1 modulators restore SWELL1-dependent insulin-sensitivity and insulin secretion in T2D and may represent a first-in-class therapeutic approach for T2D and NAFLD.
- Published
- 2021
27. Author response: The SWELL1-LRRC8 complex regulates endothelial AKT-eNOS signaling and vascular function
- Author
-
Rachel A Minerath, Megan J Riker, Chau My Ta, Robert F. Mullins, Rajan Sah, Joshua M Maurer, Ashutosh Kumar, Amber N. Stratman, Macaulay Elliot-Hudson, Susheel K. Gunasekar, Oluwaseun Adeola, Chad E. Grueter, Ahmad F. Alghanem, Urooj Fatima, Javier Abello, Chen Kang, and Litao Xie
- Subjects
biology ,Chemistry ,Enos ,Vascular function ,biology.organism_classification ,Protein kinase B ,Cell biology - Published
- 2021
28. Corrigendum to 'LRRC8A-dependent volume-regulated anion channels contribute to ischemia-induced brain injury and glutamatergic input to hippocampal neurons' [Experimental Neurology, 332(2020)113391]
- Author
-
Jing-Jing Zhou, Yi Luo, Shao-Rui Chen, Jian-Ying Shao, Rajan Sah, and Hui-Lin Pan
- Subjects
Developmental Neuroscience ,Neurology - Published
- 2022
29. Perilipin2 down-regulation in β cells impairs insulin secretion under nutritional stress and damages mitochondria
- Author
-
Siming Liu, Andrew S. Greenberg, Chen Kang, Laura Jackson, Timothy H. King, Frederick Anokye-Danso, James A. Ankrum, Rajan Sah, Brian D. Fink, Yumi Imai, Samuel B. Stephens, William I. Sivitz, Mikako Harata, Joseph A. Promes, Akansha Mishra, Gourav Bhardwaj, Rexford S. Ahima, Brian T. O’Neill, and Stefan Strack
- Subjects
endocrine system ,medicine.medical_specialty ,biology ,Chemistry ,Perilipin 2 ,Oxidative phosphorylation ,Mitochondrion ,In vitro ,Palmitic acid ,chemistry.chemical_compound ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,Lipid droplet ,medicine ,biology.protein ,Fragmentation (cell biology) - Abstract
Perilipin 2 (PLIN2) is the lipid droplet (LD) protein in β cells that increases under nutritional stress. Down-regulation of PLIN2 is often sufficient to reduce LD accumulation. To determine whether PLIN2 positively or negatively affects β cell function under nutritional stress, PLIN2 was down-regulated in mouse β cells, INS1 cells, and human islet cells. β cell specific deletion of PLIN2 in mice on a high fat diet reduced glucose-stimulated insulin secretion (GSIS) in vivo and in vitro. Down-regulation of PLIN2 in INS1 cells blunted GSIS after 24 h incubation with 0.2 mM palmitic acids. Down-regulation of PLIN2 in human pseudoislets cultured at 5.6 mM glucose impaired both phases of GSIS, indicating that PLIN2 is critical for GSIS. Down-regulation of PLIN2 decreased specific OXPHOS proteins in all three models and reduced oxygen consumption rates in INS1 cells and mouse islets. Moreover, we found that PLIN2 deficient INS1 cells increased the distribution of a fluorescent oleic acid analog to mitochondria and showed signs of mitochondrial stress as indicated by susceptibility to fragmentation and alterations of acyl-carnitines and glucose metabolites. Collectively, PLIN2 in β cells have an important role in preserving insulin secretion, β cell metabolism and mitochondrial function under nutritional stress.
- Published
- 2020
30. ORAI1 and ORAI2 modulate murine neutrophil calcium signaling, cellular activation, and host defense
- Author
-
Rajan Sah, Celeste L. Cummings, Madeline Pashos, Clifford A. Lowell, Eric Tycksen, Ryan Johnson, Helen J. McBride, Regina A. Clemens, Chen Kang, Derayvia Grimes, and Georgia R. Sampedro
- Subjects
0301 basic medicine ,Male ,ORAI1 Protein ,Neutrophils ,1.1 Normal biological development and functioning ,ORAI2 Protein ,chemistry.chemical_element ,Calcium ,Inbred C57BL ,calcium signaling ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Underpinning research ,Animals ,2.1 Biological and endogenous factors ,Calcium Signaling ,Aetiology ,Calcium signaling ,Membrane potential ,Leukotriene ,Multidisciplinary ,ORAI1 ,Endoplasmic reticulum ,Inflammatory and immune system ,Degranulation ,neutrophil ,store-operated calcium entry ,Biological Sciences ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Store-operated calcium entry ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,chemistry ,030220 oncology & carcinogenesis ,Female ,membrane potential - Abstract
Calcium signals are initiated in immune cells by the process of store-operated calcium entry (SOCE), where receptor activation triggers transient calcium release from the endoplasmic reticulum, followed by opening of plasma-membrane calcium-release activated calcium (CRAC) channels. ORAI1, ORAI2, and ORAI3 are known to comprise the CRAC channel; however, the contributions of individual isoforms to neutrophil function are not well understood. Here, we show that loss of ORAI1 partially decreases calcium influx, while loss of both ORAI1 and ORAI2 completely abolishes SOCE. In other immune-cell types, loss of ORAI2 enhances SOCE. In contrast, we find that ORAI2-deficient neutrophils display decreased calcium influx, which is correlated with measurable differences in the regulation of neutrophil membrane potential via KCa3.1. Decreased SOCE in ORAI1-, ORAI2-, and ORAI1/2-deficient neutrophils impairs multiple neutrophil functions, including phagocytosis, degranulation, leukotriene, and reactive oxygen species (ROS) production, rendering ORAI1/2-deficient mice highly susceptible to staphylococcal infection. This study demonstrates that ORAI1 and ORAI2 are the primary components of the neutrophil CRAC channel and identifies subpopulations of neutrophils where cell-membrane potential functions as a rheostat to modulate the SOCE response. These findings have implications for mechanisms that modulate neutrophil function during infection, acute and chronic inflammatory conditions, and cancer.
- Published
- 2020
31. SWELL1 regulates skeletal muscle cell size, intracellular signaling, adiposity and glucose metabolism
- Author
-
E. Dale Abel, Chau My Ta, Rajan Sah, Rachel A Minerath, Joshua M Maurer, Ashutosh Kumar, Chad E. Grueter, Gretchen A. Meyer, Susheel K. Gunasekar, Antentor Othrell Hinton, Litao Xie, and Karen Shen
- Subjects
0301 basic medicine ,Male ,Mouse ,QH301-705.5 ,medicine.medical_treatment ,growth ,Science ,Cell ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Myocyte ,Animals ,Biology (General) ,Muscle, Skeletal ,VRAC ,knock-out mouse ,Ion channel ,Adiposity ,Cell Size ,Muscle Cells ,General Immunology and Microbiology ,biology ,Chemistry ,General Neuroscience ,Insulin ,Skeletal muscle ,Membrane Proteins ,General Medicine ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Glucose ,Knockout mouse ,ion channel ,biology.protein ,Medicine ,Female ,GRB2 ,030217 neurology & neurosurgery ,Intracellular ,Signal Transduction ,Research Article - Abstract
Maintenance of skeletal muscle is beneficial in obesity and Type 2 diabetes. Mechanical stimulation can regulate skeletal muscle differentiation, growth and metabolism; however, the molecular mechanosensor remains unknown. Here, we show that SWELL1 (Lrrc8a) functionally encodes a swell-activated anion channel that regulates PI3K-AKT, ERK1/2, mTOR signaling, muscle differentiation, myoblast fusion, cellular oxygen consumption, and glycolysis in skeletal muscle cells. LRRC8A over-expression in Lrrc8a KO myotubes boosts PI3K-AKT-mTOR signaling to supra-normal levels and fully rescues myotube formation. Skeletal muscle-targeted Lrrc8a KO mice have smaller myofibers, generate less force ex vivo, and exhibit reduced exercise endurance, associated with increased adiposity under basal conditions, and glucose intolerance and insulin resistance when raised on a high-fat diet, compared to wild-type (WT) mice. These results reveal that the LRRC8 complex regulates insulin-PI3K-AKT-mTOR signaling in skeletal muscle to influence skeletal muscle differentiation in vitro and skeletal myofiber size, muscle function, adiposity and systemic metabolism in vivo., eLife digest Skeletal muscles – the force-generating tissue attached to bones – must maintain their mass and health for the body to work properly. It is therefore necessary to understand how an organism can regulate the way skeletal muscles form, grow and heal. A multitude of factors can control how muscles form, including mechanical signals. The molecules that can sense these mechanical stimuli, however, remain unknown. Mechanoresponsive ion channels provide possible candidates for these molecular sensors. These proteins are studded through the cell membranes, where they can respond to mechanical changes by opening and allowing the flow of ions in and out of a cell, or by changing interactions with other proteins. The SWELL1 protein is a component of an ion channel known as VRAC, which potentially responds to mechanical stimuli. This channel is associated with many biological processes such as cells multiplying, migrating, growing and dying, but it is still unclear how. Here, Kumar et al. first tested whether SWELL1 controls how skeletal muscle precursors mature into their differentiated and functional form. These experiments showed that SWELL1 regulates this differentiation process under the influence of the hormone insulin, as well as mechanical signals such as cell stretching. In addition, this work revealed that SWELL1 relies on an adaptor molecule called GRB2 to relay these signals in the cell. Next, Kumar et al. genetically engineered mice lacking SWELL1 only in skeletal muscle. These animals had smaller muscle cells, as well as muscles that were weaker and less enduring. When raised on a high-calorie diet, the mutant mice also got more obese and developed resistance to insulin, which is an important step driving obesity-induced diabetes. Together, these findings show that SWELL1 helps to regulate the formation and function of muscle cells, and highlights how an ion channel participates in these processes. Healthy muscles are key for overall wellbeing, as they also protect against obesity and obesity-related conditions such as type 2 diabetes or nonalcoholic fatty liver disease. This suggests that targeting SWELL1 could prove advantageous in a clinical setting.
- Published
- 2020
32. Author response: SWELL1 regulates skeletal muscle cell size, intracellular signaling, adiposity and glucose metabolism
- Author
-
E. Dale Abel, Litao Xie, Rajan Sah, Gretchen A. Meyer, Rachel A Minerath, Karen Shen, Antentor Othrell Hinton, Joshua M Maurer, Chau My Ta, Susheel K. Gunasekar, Ashutosh Kumar, and Chad E. Grueter
- Subjects
Skeletal muscle cell ,Chemistry ,Carbohydrate metabolism ,Intracellular ,Cell biology - Published
- 2020
33. SWELL1-LRRC8 complex regulates skeletal muscle cell size, intracellular signalling, adiposity and glucose metabolism
- Author
-
Rajan Sah, Antentor J. Hinton, E. Dale Abel, Gretchen A. Meyer, Litao Xie, Rachel A Minerath, Joshua M Maurer, Susheel K. Gunasekar, Karen Shen, Chad E. Grueter, Chau My Ta, and Ashutosh Kumar
- Subjects
Myoblast fusion ,medicine.anatomical_structure ,Insulin resistance ,Myogenesis ,Chemistry ,medicine ,Skeletal muscle ,Myocyte ,Glycolysis ,Carbohydrate metabolism ,medicine.disease ,Ex vivo ,Cell biology - Abstract
Maintenance of skeletal muscle is beneficial in obesity and Type 2 diabetes. Mechanical stimulation can regulate skeletal muscle differentiation, growth and metabolism, however the molecular mechanosensor remains unknown. Here, we show that SWELL1 (LRRC8a) functionally encodes a swell-activated anion channel that regulates PI3K-AKT, ERK1/2, mTOR signaling, muscle differentiation, myoblast fusion, cellular oxygen consumption, and glycolysis in skeletal muscle cells. SWELL1 over-expression in SWELL1 KO myotubes boosts PI3K-AKT-mTOR signaling to supra-normal levels and fully rescues myotube formation. Skeletal muscle targeted SWELL1 KO mice have smaller myofibers, generate less forceex vivo, and exhibit reduced exercise endurance, associated with increased adiposity under basal conditions, and glucose intolerance and insulin resistance when raised on a high-fat diet, compared to WT mice. These results reveal that the SWELL1-LRRC8 complex regulates insulin-PI3K-AKT-mTOR signalling in skeletal muscle to influence skeletal muscle differentiationin vitroand skeletal myofiber size, muscle function, adiposity and systemic metabolismin vivo.
- Published
- 2020
34. LRRC8A-dependent volume-regulated anion channels contribute to ischemia-induced brain injury and glutamatergic input to hippocampal neurons
- Author
-
Yi Luo, Shao Rui Chen, Hui Lin Pan, Rajan Sah, Jing Jing Zhou, and Jian Ying Shao
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Patch-Clamp Techniques ,Excitotoxicity ,Hippocampus ,Glutamic Acid ,Hippocampal formation ,medicine.disease_cause ,Article ,Ion Channels ,Brain ischemia ,Nestin ,03 medical and health sciences ,Glutamatergic ,Mice ,0302 clinical medicine ,Developmental Neuroscience ,Internal medicine ,medicine ,Animals ,Hypoxia ,CA1 Region, Hippocampal ,Ischemic Stroke ,Mice, Knockout ,Neurons ,Behavior, Animal ,Chemistry ,Glutamate receptor ,Excitatory Postsynaptic Potentials ,Membrane Proteins ,Infarction, Middle Cerebral Artery ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Glucose ,nervous system ,Neurology ,Excitatory postsynaptic potential ,NMDA receptor ,030217 neurology & neurosurgery - Abstract
Volume-regulated anion channels (VRACs) are critically involved in regulating cell volume, and leucine-rich repeat-containing protein 8A (LRRC8A, SWELL1) is an obligatory subunit of VRACs. Cell swelling occurs early after brain ischemia, but it is unclear whether neuronal LRRC8a contributes to ischemia-induced glutamate release and brain injury. We found that Lrrc8a conditional knockout (Lrrc8a-cKO) mice produced by crossing NestinCre+/− with Lrrc8aflox+/+ mice died 7–8 weeks of age, indicating an essential role of neuronal LRRC8A for survival. Middle cerebral artery occlusion (MCAO) caused an early increase in LRRC8A protein levels in the hippocampus in wild-type (WT) mice. Whole-cell patch-clamp recording in brain slices revealed that oxygen-glucose deprivation significantly increased the amplitude of VRAC currents in hippocampal CA1 neurons in WT but not in Lrrc8a-cKO mice. Hypotonicity increased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in hippocampal CA1 neurons in WT mice, and this was abolished by DCPIB, a VRAC blocker. But in Lrrc8a-cKO mice, hypotonic solution had no effect on the frequency of sEPSCs in these neurons. Furthermore, the brain infarct volume and neurological severity score induced by MCAO were significantly lower in Lrrc8a-cKO mice than in WT mice. In addition, MCAO-induced increases in cleaved caspase-3 and calpain activity, two biochemical markers of neuronal apoptosis and death, in brain tissues were significantly attenuated in Lrrc8a-cKO mice compared with WT mice. These new findings indicate that cerebral ischemia increases neuronal LRRC8A-dependent VRAC activity and that VRACs contribute to increased glutamatergic input to hippocampal neurons and brain injury caused by ischemic stroke.
- Published
- 2020
35. SWELL1 is a glucose sensor regulating β-cell excitability and systemic glycaemia
- Author
-
Yanhui Zhang, Saachi Pai, Ashutosh Kumar, Litao Xie, Susheel K. Gunasekar, Samuel B. Stephens, Andrew W. Norris, Anil Mishra, Rajan Sah, Yiwen Gao, and Chen Kang
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,medicine.medical_specialty ,Vesicle fusion ,medicine.medical_treatment ,Science ,General Physics and Astronomy ,chemistry.chemical_element ,Mice, Transgenic ,Calcium ,General Biochemistry, Genetics and Molecular Biology ,Calcium in biology ,Article ,03 medical and health sciences ,Islets of Langerhans ,Internal medicine ,Cell Line, Tumor ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Animals ,Humans ,Insulin ,lcsh:Science ,Calcium signaling ,Mice, Knockout ,Multidisciplinary ,Voltage-dependent calcium channel ,Membrane Proteins ,Depolarization ,General Chemistry ,030104 developmental biology ,Endocrinology ,Glucose ,chemistry ,Tonicity ,Female ,lcsh:Q ,Calcium Channels ,CRISPR-Cas Systems - Abstract
Insulin secretion is initiated by activation of voltage-gated Ca2+ channels (VGCC) to trigger Ca2+-mediated insulin vesicle fusion with the β-cell plasma membrane. The firing of VGCC requires β-cell membrane depolarization, which is regulated by a balance of depolarizing and hyperpolarizing ionic currents. Here, we show that SWELL1 mediates a swell-activated, depolarizing chloride current (ICl,SWELL) in both murine and human β-cells. Hypotonic and glucose-stimulated β-cell swelling activates SWELL1-mediated ICl,SWELL and this contributes to membrane depolarization and activation of VGCC-dependent intracellular calcium signaling. SWELL1 depletion in MIN6 cells and islets significantly impairs glucose-stimulated insulin secretion. Tamoxifen-inducible β-cell-targeted Swell1 KO mice have normal fasting serum glucose and insulin levels but impaired glucose-stimulated insulin secretion and glucose tolerance; and this is further exacerbated in mild obesity. Our results reveal that β-cell SWELL1 modulates insulin secretion and systemic glycaemia by linking glucose-mediated β-cell swelling to membrane depolarization and activation of VGCC-triggered calcium signaling., Insulin secretion by β-cells is stimulated by glucose and is dependent on the induction of β-cell membrane depolarization, mainly driven by the closure of KATP channels, which in turn promotes voltage-gated Ca2+ channel opening. Here Kang et al. show that the volume-regulated anion channel, SWELL1, is involved in glucose-stimulated calcium increase and insulin secretion.
- Published
- 2018
36. Resident cardiac macrophages mediate adaptive myocardial remodeling
- Author
-
W. Tom Stump, James A. J. Fitzpatrick, Geetika Bajpai, Attila Kovacs, Jamison Leid, Sachio Morimoto, Jay Mohan, Inessa Lokshina, Michael J. Greenberg, Shuchi Guo, Benjamin Kopecky, Daniel Kreisel, Lixia Du, Laura Ewald, Slava Epelman, Guoshuai Feng, Carla J. Weinheimer, Nicole R. Wong, Max R. Fisher, Hannah Luehmann, Rajan Sah, Kory J. Lavine, Oleksandr Dmytrenko, Nikhil R. Patel, Jessica M. Nigro, Andrea L. Bredemeyer, Hongzhen Hu, Peter O. Bayguinov, Yongjian Liu, and Lauren Bell
- Subjects
Cardiomyopathy, Dilated ,CCR2 ,medicine.medical_treatment ,Immunology ,Population ,Biology ,Article ,Focal adhesion ,Mice ,Chemokine receptor ,Troponin T ,medicine ,Animals ,Humans ,Immunology and Allergy ,Macrophage ,education ,Ventricular remodeling ,education.field_of_study ,Ventricular Remodeling ,Macrophages ,Myocardium ,Growth factor ,Macrophage Activation ,medicine.disease ,Mice, Mutant Strains ,Cell biology ,Mice, Inbred C57BL ,Infectious Diseases ,Heart failure ,Mutation - Abstract
Cardiac macrophages represent a heterogeneous cell population with distinct origins, dynamics, and functions. Recent studies have revealed that C-C Chemokine Receptor 2 positive (CCR2(+)) macrophages derived from infiltrating monocytes regulate myocardial inflammation and heart failure pathogenesis. Comparatively little is known about the functions of tissue resident (CCR2(−)) macrophages. Herein, we identified an essential role for CCR2(−) macrophages in the chronically failing heart. Depletion of CCR2(−) macrophages in mice with dilated cardiomyopathy accelerated mortality and impaired ventricular remodeling and coronary angiogenesis, adaptive changes necessary to maintain cardiac output in the setting of reduced cardiac contractility. Mechanistically, CCR2(−) macrophages interacted with neighboring cardiomyocytes via focal adhesion complexes and were activated in response to mechanical stretch through a transient receptor potential vanilloid 4 (TRPV4) dependent pathway that controlled growth factor expression. These findings establish a role for tissue resident macrophages in adaptive cardiac remodeling and implicate mechanical sensing in cardiac macrophage activation.
- Published
- 2021
37. Mechanism and effects of pulsatile GABA secretion from cytosolic pools in the human beta cell
- Author
-
Matthew W. Becker, Judith Molina, Joana Almaça, Robert M. Dolan, Rayner Rodriguez-Diaz, Alejandro Caicedo, Rita Nano, Rajan Sah, Chen Kang, Herbert Y. Gaisano, Chiara Cianciaruso, Danusa Menegaz, Edward A. Phelps, Per Olof Berggren, D. Walker Hagan, Steinunn Baekkeskov, Petra C. Schwalie, and Fanny Lebreton
- Subjects
Pulsatile insulin ,Endocrinology, Diabetes and Metabolism ,aminobutyric-acid gaba ,essential component ,Autoimmune Disease ,Article ,pancreatic-islet cells ,Paracrine signalling ,Cytosol ,golgi membranes ,alpha-cells ,Physiology (medical) ,Insulin-Secreting Cells ,Internal Medicine ,Diabetes Mellitus ,Humans ,Homeostasis ,2.1 Biological and endogenous factors ,Secretion ,glucose-inhibition ,Aetiology ,Autocrine signalling ,gamma-Aminobutyric Acid ,Metabolic and endocrine ,geography ,geography.geographical_feature_category ,ddc:617 ,Chemistry ,Diabetes ,Glucagon secretion ,Neurosciences ,Cell Biology ,Islet ,Cell biology ,substrate-specificity ,Diabetes Mellitus, Type 1 ,nervous system ,Diabetes Mellitus, Type 2 ,synaptic-like microvesicles ,glucagon-secretion ,Beta cell ,Type 2 ,insulin-secretion ,Subcellular Fractions ,Type 1 - Abstract
Pancreatic beta cells synthesize and secrete the neurotransmitter GABA (gamma-aminobutyric acid) as a paracrine and autocrine signal to help regulate hormone secretion and islet homeostasis. Islet GABA release has classically been described as a secretory-vesicle-mediated event. Yet, a limitation of the hypothesized vesicular GABA release from islets is the lack of expression of a vesicular GABA transporter in beta cells. Consequentially, GABA accumulates in the cytosol. Here, we provide evidence that the human beta cell effluxes GABA from a cytosolic pool in a pulsatile manner, imposing a synchronizing rhythm on pulsatile insulin secretion. The volume regulatory anion channel, functionally encoded by LRRC8A or Swell1, is critical for pulsatile GABA secretion. GABA content in beta cells is depleted and secretion is disrupted in islets from patients with type 1 and type 2 diabetes, suggesting that loss of GABA as a synchronizing signal for hormone output may correlate with diabetes pathogenesis.
- Published
- 2019
38. Transient Receptor Potential channels (version 2019.4) in the IUPHAR/BPS Guide to Pharmacology Database
- Author
-
Elena Oancea, Paul G. DeCaen, Ingrid Carvacho, Boyi Liu, Rajan Sah, Sven E. Jordt, Antonio Riccio, Lu Fan, Kotdaji Ha, Julia F. Doerner, David D. McKemy, Jinbin Tian, Michael X. Zhu, Nathaniel T. Blair, David Julius, Haoxing Xu, Dan Tong, Markus Delling, Kristopher T. Kahle, Dipayan Chaudhuri, Stephanie C. Stotz, Grzegorz Owsianik, Long Jun Wu, Bernd Nilius, Xiaoli Zhang, Lixia Yue, Charlotte Van den Eynde, Joris Vriens, and David E. Clapham
- Subjects
Transient receptor potential channel ,Chemistry ,Neuroscience - Abstract
The TRP superfamily of channels (nomenclature as agreed by NC-IUPHAR [145, 915]), whose founder member is the Drosophila Trp channel, exists in mammals as six families; TRPC, TRPM, TRPV, TRPA, TRPP and TRPML based on amino acid homologies. TRP subunits contain six putative transmembrane domains and assemble as homo- or hetero-tetramers to form cation selective channels with diverse modes of activation and varied permeation properties (reviewed by [630]). Established, or potential, physiological functions of the individual members of the TRP families are discussed in detail in the recommended reviews and in a number of books [344, 589, 979, 216]. The established, or potential, involvement of TRP channels in disease is reviewed in [384, 588] and [591], together with a special edition of Biochemica et Biophysica Acta on the subject [588]. Additional disease related reviews, for pain [542], stroke [967], sensation and inflammation [843], itch [109], and airway disease [261, 896], are available. The pharmacology of most TRP channels has been advanced in recent years. Broad spectrum agents are listed in the tables along with more selective, or recently recognised, ligands that are flagged by the inclusion of a primary reference. See Rubaiy (2019) for a review of pharmacological tools for TRPC1/C4/C5 channels [692]. Most TRP channels are regulated by phosphoinostides such as PtIns(4,5)P2 although the effects reported are often complex, occasionally contradictory, and likely to be dependent upon experimental conditions, such as intracellular ATP levels (reviewed by [862, 592, 689]). Such regulation is generally not included in the tables.When thermosensitivity is mentioned, it refers specifically to a high Q10 of gating, often in the range of 10-30, but does not necessarily imply that the channel's function is to act as a 'hot' or 'cold' sensor. In general, the search for TRP activators has led to many claims for temperature sensing, mechanosensation, and lipid sensing. All proteins are of course sensitive to energies of binding, mechanical force, and temperature, but the issue is whether the proposed input is within a physiologically relevant range resulting in a response. TRPA (ankyrin) familyTRPA1 is the sole mammalian member of this group (reviewed by [246]). TRPA1 activation of sensory neurons contribute to nociception [356, 763, 516]. Pungent chemicals such as mustard oil (AITC), allicin, and cinnamaldehyde activate TRPA1 by modification of free thiol groups of cysteine side chains, especially those located in its amino terminus [491, 47, 311, 493]. Alkenals with α, β-unsaturated bonds, such as propenal (acrolein), butenal (crotylaldehyde), and 2-pentenal can react with free thiols via Michael addition and can activate TRPA1. However, potency appears to weaken as carbon chain length increases [21, 47]. Covalent modification leads to sustained activation of TRPA1. Chemicals including carvacrol, menthol, and local anesthetics reversibly activate TRPA1 by non-covalent binding [364, 438, 923, 922]. TRPA1 is not mechanosensitive under physiological conditions, but can be activated by cold temperatures [365, 175]. The electron cryo-EM structure of TRPA1 [639] indicates that it is a 6-TM homotetramer. Each subunit of the channel contains two short ‘pore helices’ pointing into the ion selectivity filter, which is big enough to allow permeation of partially hydrated Ca2+ ions. TRPC (canonical) familyMembers of the TRPC subfamily (reviewed by [239, 673, 14, 4, 79, 382, 638, 55]) fall into the subgroups outlined below. TRPC2 is a pseudogene in humans. It is generally accepted that all TRPC channels are activated downstream of Gq/11-coupled receptors, or receptor tyrosine kinases (reviewed by [661, 814, 915]). A comprehensive listing of G-protein coupled receptors that activate TRPC channels is given in [4]. Hetero-oligomeric complexes of TRPC channels and their association with proteins to form signalling complexes are detailed in [14] and [383]. TRPC channels have frequently been proposed to act as store-operated channels (SOCs) (or compenents of mulimeric complexes that form SOCs), activated by depletion of intracellular calcium stores (reviewed by [640, 14, 665, 703, 954, 132, 626, 51, 133]). However, the weight of the evidence is that they are not directly gated by conventional store-operated mechanisms, as established for Stim-gated Orai channels. TRPC channels are not mechanically gated in physiologically relevant ranges of force. All members of the TRPC family are blocked by 2-APB and SKF96365 [295, 294]. Activation of TRPC channels by lipids is discussed by [55]. Important progress has been recently made in TRPC pharmacology [692, 529, 372, 87]. TRPC channels regulate a variety of physiological functions and are implicated in many human diseases [248, 56, 759, 879]. TRPC1/C4/C5 subgroup TRPC1 alone may not form a functional ion channel [191]. TRPC4/C5 may be distinguished from other TRP channels by their potentiation by micromolar concentrations of La3+. TRPC2 is a pseudogene in humans, but in other mammals appears to be an ion channel localized to microvilli of the vomeronasal organ. It is required for normal sexual behavior in response to pheromones in mice. It may also function in the main olfactory epithelia in mice [951, 625, 624, 952, 462, 988, 947].TRPC3/C6/C7 subgroup All members are activated by diacylglycerol independent of protein kinase C stimulation [295].TRPM (melastatin) familyMembers of the TRPM subfamily (reviewed by [230, 294, 640, 978]) fall into the five subgroups outlined below. TRPM1/M3 subgroupIn darkness, glutamate released by the photoreceptors and ON-bipolar cells binds to the metabotropic glutamate receptor 6 , leading to activation of Go . This results in the closure of TRPM1. When the photoreceptors are stimulated by light, glutamate release is reduced, and TRPM1 channels are more active, resulting in cell membrane depolarization. Human TRPM1 mutations are associated with congenital stationary night blindness (CSNB), whose patients lack rod function. TRPM1 is also found melanocytes. Isoforms of TRPM1 may present in melanocytes, melanoma, brain, and retina. In melanoma cells, TRPM1 is prevalent in highly dynamic intracellular vesicular structures [341, 609]. TRPM3 (reviewed by [615]) exists as multiple splice variants which differ significantly in their biophysical properties. TRPM3 is expressed in somatosensory neurons and may be important in development of heat hyperalgesia during inflammation (see review [803]). TRPM3 is frequently coexpressed with TRPA1 and TRPV1 in these neurons. TRPM3 is expressed in pancreatic beta cells as well as brain, pituitary gland, eye, kidney, and adipose tissue [614, 802]. TRPM3 may contribute to the detection of noxious heat [870].TRPM2TRPM2 is activated under conditions of oxidative stress (respiratory burst of phagocytic cells) and ischemic conditions. However, the direct activators are ADPR(P) and calcium. As for many ion channels, PIP2 must also be present (reviewed by [935]). Numerous splice variants of TRPM2 exist which differ in their activation mechanisms [200]. The C-terminal domain contains a TRP motif, a coiled-coil region, and an enzymatic NUDT9 homologous domain. TRPM2 appears not to be activated by NAD, NAAD, or NAADP, but is directly activated by ADPRP (adenosine-5'-O-disphosphoribose phosphate) [827]. TRPM2 is involved in warmth sensation [724], and contributes to neurological diseases [61]. Recent study shows that 2'-deoxy-ADPR is an endogenous TRPM2 superagonist [231]. TRPM4/5 subgroupTRPM4 and TRPM5 have the distinction within all TRP channels of being impermeable to Ca2+ [915]. A splice variant of TRPM4 (i.e.TRPM4b) and TRPM5 are molecular candidates for endogenous calcium-activated cation (CAN) channels [278]. TRPM4 is active in the late phase of repolarization of the cardiac ventricular action potential. TRPM4 deletion or knockout enhances beta adrenergic-mediated inotropy [507]. Mutations are associated with conduction defects [347, 507, 753]. TRPM4 has been shown to be an important regulator of Ca2+ entry in to mast cells [847] and dendritic cell migration [39]. TRPM5 in taste receptor cells of the tongue appears essential for the transduction of sweet, amino acid and bitter stimuli [460] TRPM5 contributes to the slow afterdepolarization of layer 5 neurons in mouse prefrontal cortex [439]. Both TRPM4 and TRPM5 are required transduction of taste stimuli [206].TRPM6/7 subgroupTRPM6 and 7 combine channel and enzymatic activities (‘chanzymes’). These channels have the unusual property of permeation by divalent (Ca2+, Mg2+, Zn2+) and monovalent cations, high single channel conductances, but overall extremely small inward conductance when expressed to the plasma membrane. They are inhibited by internal Mg2+ at ~0.6 mM, around the free level of Mg2+ in cells. Whether they contribute to Mg2+ homeostasis is a contentious issue. When either gene is deleted in mice, the result is embryonic lethality. The C-terminal kinase region is cleaved under unknown stimuli, and the kinase phosphorylates nuclear histones. TRPM7 is responsible for oxidant- induced Zn2+ release from intracellular vesicles [3] and contributes to intestinal mineral absorption essential for postnatal survival [532]. TRPM8Is a channel activated by cooling and pharmacological agents evoking a ‘cool’ sensation and participates in the thermosensation of cold temperatures [50, 147, 186] reviewed by [864, 481, 391, 556]. TRPML (mucolipin) familyThe TRPML family [676, 964, 670, 926, 156] consists of three mammalian members (TRPML1-3). TRPML channels are probably restricted to intracellular vesicles and mutations in the gene (MCOLN1) encoding TRPML1 (mucolipin-1) cause the neurodegenerative disorder mucolipidosis type IV (MLIV) in man. TRPML1 is a cation selective ion channel that is important for sorting/transport of endosomes in the late endocytotic pathway and specifically, fission from late endosome-lysosome hybrid vesicles and lysosomal exocytosis [704]. TRPML2 and TRPML3 show increased channel activity in low extracellular sodium and are activated by similar small molecules [270]. A naturally occurring gain of function mutation in TRPML3 (i.e. A419P) results in the varitint waddler (Va) mouse phenotype (reviewed by [676, 593]). TRPP (polycystin) familyThe TRPP family (reviewed by [179, 177, 252, 905, 320]) or PKD2 family is comprised of PKD2 (PC2), PKD2L1 (PC2L1), PKD2L2 (PC2L2), which have been renamed TRPP1, TRPP2 and TRPP3, respectively [915]. It should also be noted that the nomenclature of PC2 was TRPP2 in old literature. However, PC2 has been uniformed to be called TRPP2 [293]. PKD2 family channels are clearly distinct from the PKD1 family, whose function is unknown. PKD1 and PKD2 form a hetero-oligomeric complex with a 1:3 ratio. [775]. Although still being sorted out, TRPP family members appear to be 6TM spanning nonselective cation channels. TRPV (vanilloid) familyMembers of the TRPV family (reviewed by [849]) can broadly be divided into the non-selective cation channels, TRPV1-4 and the more calcium selective channels TRPV5 and TRPV6.TRPV1-V4 subfamilyTRPV1 is involved in the development of thermal hyperalgesia following inflammation and may contribute to the detection of noxius heat (reviewed by [660, 756, 786]). Numerous splice variants of TRPV1 have been described, some of which modulate the activity of TRPV1, or act in a dominant negative manner when co-expressed with TRPV1 [722]. The pharmacology of TRPV1 channels is discussed in detail in [280] and [868]. TRPV2 is probably not a thermosensor in man [635], but has recently been implicated in innate immunity [469]. TRPV3 and TRPV4 are both thermosensitive. There are claims that TRPV4 is also mechanosensitive, but this has not been established to be within a physiological range in a native environment [106, 454].TRPV5/V6 subfamily TRPV5 and TRPV6 are highly expressed in placenta, bone, and kidney. Under physiological conditions, TRPV5 and TRPV6 are calcium selective channels involved in the absorption and reabsorption of calcium across intestinal and kidney tubule epithelia (reviewed by [901, 168, 558, 227]).
- Published
- 2019
39. Role of diabetes and insulin use in the risk of stroke and acute myocardial infarction in patients with atrial fibrillation: A Medicare analysis
- Author
-
Mary Vaughan Sarrazin, Rajan Sah, Michael C. Giudici, Geoffrey D. Barnes, Oluwaseun Adeola, Amgad Mentias, Deborah A. Levine, Konstantinos C. Siontis, Ghanshyam Palamaner Subash Shantha, and Bharat Narasimhan
- Subjects
Male ,Risk ,medicine.medical_specialty ,Time Factors ,medicine.drug_class ,Diabetic Cardiomyopathies ,medicine.medical_treatment ,Myocardial Infarction ,030204 cardiovascular system & hematology ,Medicare ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,Atrial Fibrillation ,medicine ,Diabetes Mellitus ,Humans ,Hypoglycemic Agents ,Insulin ,030212 general & internal medicine ,Myocardial infarction ,Stroke ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,business.industry ,Proportional hazards model ,Anticoagulant ,Anticoagulants ,Atrial fibrillation ,medicine.disease ,INSULIN USE ,United States ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Diabetic Angiopathies - Abstract
BACKGROUND: Atrial fibrillation (AF) is associated with elevated risk for ischemic stroke and myocardial infarction (MI). The aim of the study is to assess the role of insulin use on the risk of stroke and MI in AF patients with diabetes. METHODS: We identified Medicare beneficiaries with new AF in 2011-2013. Primary outcomes were ischemic stroke and MI. Multivariate Cox regression models were used to assess the association between AF and time to stroke and MI. We adjusted for anticoagulant as a time-dependent covariate. RESULTS: Out of 798,592 AF patients, 53212 (6.7%) were insulin-requiring diabetics (IRD), 250214 (31.3%) were non-insulin requiring diabetics (NIRD) and 495166 (62%) were non-diabetics (ND). IRD had a higher risk of stroke when compared to NIRD (adjusted HR: 1.15, 95% CI 1.10-1.21) and ND (aHR 1.24, 95% CI 1.18-1.31) (P
- Published
- 2018
40. Old tools, new applications: Use of environmental bacteriophages for typhoid surveillance and evaluating vaccine impact.
- Author
-
Yogesh Hooda, Shuborno Islam, Rathin Kabiraj, Hafizur Rahman, Himadree Sarkar, Kesia E da Silva, Rajan Saha Raju, Stephen P Luby, Jason R Andrews, Samir K Saha, and Senjuti Saha
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Typhoid-conjugate vaccines (TCVs) provide an opportunity to reduce the burden of typhoid fever, caused by Salmonella Typhi, in endemic areas. As policymakers design vaccination strategies, accurate and high-resolution data on disease burden is crucial. However, traditional blood culture-based surveillance is resource-extensive, prohibiting its large-scale and sustainable implementation. Salmonella Typhi is a water-borne pathogen, and here, we tested the potential of Typhi-specific bacteriophage surveillance in surface water bodies as a low-cost tool to identify where Salmonella Typhi circulates in the environment. In 2021, water samples were collected and tested for the presence of Salmonella Typhi bacteriophages at two sites in Bangladesh: urban capital city, Dhaka, and a rural district, Mirzapur. Salmonella Typhi-specific bacteriophages were detected in 66 of 211 (31%) environmental samples in Dhaka, in comparison to 3 of 92 (3%) environmental samples from Mirzapur. In the same year, 4,620 blood cultures at the two largest pediatric hospitals of Dhaka yielded 215 (5%) culture-confirmed typhoid cases, and 3,788 blood cultures in the largest hospital of Mirzapur yielded 2 (0.05%) cases. 75% (52/69) of positive phage samples were collected from sewage. All isolated phages were tested against a panel of isolates from different Salmonella Typhi genotypes circulating in Bangladesh and were found to exhibit a diverse killing spectrum, indicating that diverse bacteriophages were isolated. These results suggest an association between the presence of Typhi-specific phages in the environment and the burden of typhoid fever, and the potential of utilizing environmental phage surveillance as a low-cost tool to assist policy decisions on typhoid control.
- Published
- 2024
- Full Text
- View/download PDF
41. Induction of adipose and hepatic SWELL1 expression is required for maintaining systemic insulin-sensitivity in obesity
- Author
-
Susheel K. Gunasekar, Anil Mishra, Litao Xie, Rajan Sah, Lei Cao, and Yanhui Zhang
- Subjects
0301 basic medicine ,Anions ,medicine.medical_specialty ,medicine.medical_treatment ,Biophysics ,Insulins ,Adipose tissue ,Type 2 diabetes ,Diet, High-Fat ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Insulin resistance ,Adipocyte ,Diabetes mellitus ,Internal medicine ,medicine ,Adipocytes ,Glucose homeostasis ,Animals ,Homeostasis ,Obesity ,RNA, Small Interfering ,biology ,business.industry ,Insulin ,Membrane Proteins ,Dependovirus ,medicine.disease ,Article Addendum ,Mice, Inbred C57BL ,Insulin receptor ,030104 developmental biology ,Endocrinology ,Glucose ,chemistry ,Liver ,biology.protein ,Insulin Resistance ,business ,Signal Transduction - Abstract
Obesity is associated with a loss of insulin-sensitivity and systemic dysglycemia, resulting in Type 2 diabetes, however the molecular mechanisms underlying this association are unclear. Through adipocyte patch-clamp studies, we recently showed that SWELL1 is required for the Volume-Regulated Anion Current (VRAC) in adipocytes and that SWELL1-mediated VRAC is activated by both mechanical and pathophysiological adipocyte expansion. We also demonstrated that adipocyte SWELL1 is required for maintaining insulin signaling and glucose homeostasis, particularly in the setting of obesity. Here we show that SWELL1 protein expression is induced in subcutaneous fat, visceral fat and liver in the setting of obesity. Long- term AAV/rec2-shRNA mediated SWELL1 knock-down in both fat and liver are associated with increased weight gain, increased adiposity and exacerbated insulin resistance in mice raised on a high-fat diet. These data further support the notion that SWELL1 induction occurs in insulin- sensitive tissues (liver and adipose) in the setting of over-nutrition and contributes to improved systemic glycemia by supporting enhanced insulin-sensitivity.
- Published
- 2017
42. TRPM7 senses oxidative stress to release Zn
- Author
-
Sunday A, Abiria, Grigory, Krapivinsky, Rajan, Sah, Ana G, Santa-Cruz, Dipayan, Chaudhuri, Jin, Zhang, Pichet, Adstamongkonkul, Paul G, DeCaen, and David E, Clapham
- Subjects
inorganic chemicals ,Oxidative Stress ,Zinc ,HEK293 Cells ,PNAS Plus ,Embryonic Development ,Humans ,TRPM Cation Channels ,Protein Serine-Threonine Kinases ,Reactive Oxygen Species ,Transport Vesicles ,Glutathione - Abstract
TRPM7 (transient receptor potential cation channel subfamily M member 7) is required for normal organ development but also mediates anoxic neuronal death. TRPM7 contains a channel that conducts cations into the cytosol and C-terminal kinase that can phosphorylate multiple substrates. The kinase is cleaved to regulate gene expression and apoptosis. The link between TRPM7’s channel and its organismal function remains the least understood aspect of TRPM7. Here, we identify intracellular Zn2+ storage vesicles that contain the majority of TRPM7 protein. TRPM7 senses reactive oxygen species (ROS) to release Zn2+ from these vesicles. Just as the endoplasmic reticulum sequesters and releases Ca2+, we propose that these vesicles fulfill a similar function for Zn2+ and that TRPM7 coordinates fluctuations in cellular [Zn2+] and ROS during development and injury.
- Published
- 2017
43. TRPM7 senses oxidative stress to release Zn 2+ from unique intracellular vesicles
- Author
-
Rajan Sah, Sunday A. Abiria, Grigory Krapivinsky, Dipayan Chaudhuri, Paul G. DeCaen, Jin Zhang, Pichet Adstamongkonkul, Ana Santa-Cruz, and David E. Clapham
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Multidisciplinary ,Endosome ,Vesicle ,Endoplasmic reticulum ,Biology ,Divalent ,Cell biology ,03 medical and health sciences ,Transient receptor potential channel ,Cytosol ,030104 developmental biology ,chemistry ,TRPM7 ,Intracellular - Abstract
Significance TRPM7 (transient receptor potential cation channel subfamily M member 7) is required for normal organ development but also mediates anoxic neuronal death. TRPM7 contains a channel that conducts cations into the cytosol and C-terminal kinase that can phosphorylate multiple substrates. The kinase is cleaved to regulate gene expression and apoptosis. The link between TRPM7’s channel and its organismal function remains the least understood aspect of TRPM7. Here, we identify intracellular Zn 2+ storage vesicles that contain the majority of TRPM7 protein. TRPM7 senses reactive oxygen species (ROS) to release Zn 2+ from these vesicles. Just as the endoplasmic reticulum sequesters and releases Ca 2+ , we propose that these vesicles fulfill a similar function for Zn 2+ and that TRPM7 coordinates fluctuations in cellular [Zn 2+ ] and ROS during development and injury.
- Published
- 2017
44. SWELL1 is a glucose sensor required for β-cell excitability and insulin secretion
- Author
-
Samuel B. Stephens, Yanhui Zhang, Saachi Pai, Andrew W. Norris, Rajan Sah, Yiwen Gao, Litao Xie, Anil Mishra, Susheel K. Gunasekar, and Chen Kang
- Subjects
Membrane potential ,0303 health sciences ,medicine.medical_specialty ,Insulin ,medicine.medical_treatment ,Depolarization ,Biology ,Inhibitory postsynaptic potential ,Calcium in biology ,Insulin oscillation ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Excitatory postsynaptic potential ,030217 neurology & neurosurgery ,030304 developmental biology ,Calcium signaling - Abstract
Insulin secretion from the pancreatic β-cell initiated by activation of voltage-gated Ca2+ channels (VGCC) to trigger Ca2+-mediated insulin vesicle fusion with the β-cell plasma membrane. The firing of VGCC depends on the β-cell membrane potential, which is in turn mediated by the balance of depolarizing (excitatory) and hyperpolarizing (inhibitory) ionic currents1-3. While much attention has focused on inhibitory potassium currents4-10 there is little knowledge about the excitatory currents required to depolarize the β-cell, including the molecular identity of these excitatory currents3. Here we show that SWELL1 (LRRC8a) mediates a swell-activated, depolarizing chloride current (ICl,SWELL) in β-cells. Hypotonic and glucose-stimulated β-cell swelling activates SWELL1-mediated ICl,SWELL and this is required for both glucose-stimulated and hypotonic swell-mediated activation of VGCC-dependent intracellular calcium signaling in β-cells. SWELL1 KO MIN6 cells and β-cell targeted SWELL1 KO murine islets exhibit significantly impaired glucose-stimulated insulin secretion, with preserved insulin content in vitro. Tamoxifen-inducible β-cell targeted SWELL1 KO mice have normal fasting insulin levels but display markedly impaired glucose-stimulated insulin secretion. Our results reveal a physiological role for SWELL1 as a glucose sensor - linking glucose-mediated β-cell swelling to SWELL1-dependent activation of VGCC-triggered calcium signaling, and highlights SWELL1-mediated “swell-secretion” coupling as required for glucose-stimulated insulin secretion.
- Published
- 2017
- Full Text
- View/download PDF
45. The volume-regulated anion channel (LRRC8) in nodose neurons is sensitive to acidic pH
- Author
-
Yanhui Zhang, Francois M. Abboud, Rajan Sah, Gunasekar Susheel, Christopher J. Benson, Yongjun Lu, Mark W. Chapleau, and Runping Wang
- Subjects
0301 basic medicine ,Patch-Clamp Techniques ,Neurite ,Neuroprotection ,03 medical and health sciences ,Mice ,Chlorides ,Ganglia, Spinal ,Extracellular ,Animals ,Humans ,Patch clamp ,RNA, Messenger ,Ischemic Preconditioning ,Cells, Cultured ,Neurons ,Chemistry ,HEK 293 cells ,Osmolar Concentration ,Membrane Proteins ,NADPH Oxidases ,General Medicine ,Hydrogen Peroxide ,Hydrogen-Ion Concentration ,Cell biology ,030104 developmental biology ,HEK293 Cells ,Cell culture ,Apoptosis ,Acids ,Intracellular ,Research Article - Abstract
The leucine rich repeat containing protein 8A (LRRC8A), or SWELL1, is an essential component of the volume-regulated anion channel (VRAC) that is activated by cell swelling and ionic strength. We report here for the first time to our knowledge its expression in a primary cell culture of nodose ganglia neurons and its localization in the soma, neurites, and neuronal membrane. We show that this neuronal VRAC/SWELL1 senses low external pH (pHo) in addition to hypoosmolarity. A robust sustained chloride current is seen in 77% of isolated nodose neurons following brief exposures to extracellular acid pH. Its activation involves proton efflux, intracellular alkalinity, and an increase in NOX-derived H2O2. The molecular identity of both the hypoosmolarity-induced and acid pHo-conditioned VRAC as LRRC8A (SWELL1) was confirmed by Cre-flox-mediated KO, shRNA-mediated knockdown, and CRISPR/Cas9-mediated LRRC8A deletion in HEK cells and in primary nodose neuronal cultures. Activation of VRAC by low pHo reduces neuronal injury during simulated ischemia and N-methyl-D-aspartate-induced (NMDA-induced) apoptosis. These results identify the VRAC (LRRC8A) as a dual sensor of hypoosmolarity and low pHo in vagal afferent neurons and define the mechanisms of its activation and its neuroprotective potential.
- Published
- 2017
46. Isolation and Patch-Clamp of Primary Adipocytes
- Author
-
Yanhui, Zhang, Dan, Tong, Anil, Mishra, Litao, Xie, Isaac, Samuel, Jessica K, Smith, and Rajan, Sah
- Subjects
Mice ,Patch-Clamp Techniques ,Adipocytes ,Cell Culture Techniques ,Animals ,Humans ,Cell Differentiation ,Cell Separation ,Electrophysiological Phenomena - Abstract
The patch-clamp technique allows for the study of ion channel activity in the native adipocyte environment to better understand the contributions of ion channels to adipocyte signaling. Here, we describe methods for isolating primary mature adipocytes from both mouse and human white adipose tissues (subcutaneous and visceral). From the same preparation, we describe how to culture and differentiate preadipocytes isolated from the stromal vascular fraction. We then describe in detail patch-clamp methods, including both whole-cell and perforated-patch configurations.
- Published
- 2017
47. The Swell1-LRRC8 Complex Regulates Endothelial PI3K-AKT2-GRB2-eNOS Signaling and Vascular Function
- Author
-
Susheel K. Gunasekar, Robert F. Mullins, Chau Ta, Elliot-Hudson Elliot-Hudson, Oluwaseun Adeola, Yanhui Zhang, Megan J Riker, Litao Xie, Ahmad F. Alghanem, Rajan Sah, and Urooj Fatima
- Subjects
biology ,Enos ,Chemistry ,Biophysics ,biology.protein ,AKT2 ,GRB2 ,biology.organism_classification ,Vascular function ,PI3K/AKT/mTOR pathway ,Cell biology - Published
- 2019
48. Timing of Myocardial Trpm7 Deletion During Cardiogenesis Variably Disrupts Adult Ventricular Function, Conduction, and Repolarization
- Author
-
David E. Clapham, William J. Gibson, Dipayan Chaudhuri, Marjolein A.W. van den Boogert, Matteo E. Mangoni, Christopher Bates-Withers, Pietro Mesirca, Rajan Sah, William T. Pu, and Xenos Mason
- Subjects
medicine.medical_specialty ,Mice, 129 Strain ,Time Factors ,Heart block ,Action Potentials ,TRPM Cation Channels ,Biology ,Article ,TRPC1 ,Mice ,TRPC3 ,Heart Conduction System ,TRPM7 ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Ventricular Function ,Repolarization ,Myocytes, Cardiac ,Mice, Knockout ,Myocardium ,Age Factors ,medicine.disease ,Heart failure ,Knockout mouse ,Cardiology ,Electrical conduction system of the heart ,Cardiology and Cardiovascular Medicine ,Gene Deletion - Abstract
Background— Transient receptor potential ( TRP ) channels are a superfamily of broadly expressed ion channels with diverse physiological roles. TRPC1, TRPC3 , and TRPC6 are believed to contribute to cardiac hypertrophy in mouse models. Human mutations in TRPM4 have been linked to progressive familial heart block. TRPM7 is a divalent-permeant channel and kinase of unknown function, recently implicated in the pathogenesis of atrial fibrillation; however, its function in ventricular myocardium remains unexplored. Methods and Results— We generated multiple cardiac-targeted knockout mice to test the hypothesis that TRPM7 is required for normal ventricular function. Early cardiac Trpm7 deletion (before embryonic day 9; TnT / Isl1-C re) results in congestive heart failure and death by embryonic day 11.5 as a result of hypoproliferation of the compact myocardium. Remarkably, Trpm7 deletion late in cardiogenesis (about embryonic day 13; αMHC-Cre ) produces viable mice with normal adult ventricular size, function, and myocardial transcriptional profile. Trpm7 deletion at an intermediate time point results in 50% of mice developing cardiomyopathy associated with heart block, impaired repolarization, and ventricular arrhythmias. Microarray analysis reveals elevations in transcripts of hypertrophy/remodeling genes and reductions in genes important for suppressing hypertrophy ( Hdac9) and for ventricular repolarization ( Kcnd2 ) and conduction ( Hcn4 ). These transcriptional changes are accompanied by action potential prolongation and reductions in transient outward current ( I to ; Kcnd2 ). Similarly, the pacemaker current ( I f ; Hcn4 ) is suppressed in atrioventricular nodal cells, accounting for the observed heart block. Conclusions— Trpm7 is dispensable in adult ventricular myocardium under basal conditions but is critical for myocardial proliferation during early cardiogenesis. Loss of Trpm7 at an intermediate developmental time point alters the myocardial transcriptional profile in adulthood, impairing ventricular function, conduction, and repolarization.
- Published
- 2013
49. A Rare Pericardial Malignancy
- Author
-
Dennis J. Firchau, Urooj Fatima, Alan H. Stolpen, Sarika Gupta, and Rajan Sah
- Subjects
Male ,Mesothelioma ,Orthopnea ,Respiratory rate ,Sinus tachycardia ,Computed Tomography Angiography ,Biopsy ,Cardiovascular examination ,030204 cardiovascular system & hematology ,Chest pain ,Diagnosis, Differential ,Heart Neoplasms ,03 medical and health sciences ,Electrocardiography ,0302 clinical medicine ,Medicine ,Humans ,Heart Failure ,medicine.diagnostic_test ,business.industry ,Thoracic Surgery, Video-Assisted ,Stroke Volume ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,030220 oncology & carcinogenesis ,Anesthesia ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Right axis deviation ,Chest radiograph ,Pericardium ,Paroxysmal Nocturnal Dyspnea - Abstract
A 60-year-old man without significant past medical history presented with 1 month of gradually increasing dyspnea on exertion, associated with generalized fatigue and pedal edema. He denied orthopnea, paroxysmal nocturnal dyspnea, chest pain, fever, cough, or weight loss. One month before this presentation, he could walk for miles and ride a road bike without difficulty. His social history was notable for 45 pack-year smoking and intravenous drug abuse. He worked as a refuse collector and roof mechanic for 2 to 3 years. Vital signs on presentation revealed a blood pressure 97/72 mm Hg, pulse 112 beats per minute, temperature 36.9°C, and respiratory rate of 24 breaths per minute. The oxygen saturation was 98% on room air. Cardiovascular examination revealed mild jugular venous distention to 10 cm, distant heart sounds with no murmurs, an impalpable apical impulse, and 2+ bilateral lower extremity edema. Pulmonary examination was unremarkable with clear breath sounds on auscultation bilaterally. Abdominal and neurological examination was unremarkable. Laboratory testing revealed negative troponins and mildly positive D-Dimer at 0.93 μg/mL. Although N-terminal pro b-type natriuretic peptide was elevated at 1382 pg/mL, a 2-view chest radiograph showed normal cardio-mediastinal silhouette and pulmonary vasculature. An ECG on admission showed sinus tachycardia with a rate of 110 beats per minute, right axis deviation, low-voltage complexes, and T wave inversions in leads V4, V5, and V6. Based on examination findings and initial testing, a computed tomographic …
- Published
- 2016
50. SWELL1 is a regulator of adipocyte size, insulin signalling and glucose homeostasis
- Author
-
Aloysius J. Klingelhutz, Anil Mishra, Jessica K. Smith, Yanhui Zhang, Susheel K. Gunasekar, Isaac Samuel, William J. Gibson, Rajan Sah, Dan Tong, Brodie Marthaler, Chuansong Wang, Litao Xie, E. Dale Abel, Trevor P. Fidler, and Lei Cao
- Subjects
0301 basic medicine ,Male ,Time Factors ,Glucose uptake ,Membrane Potentials ,chemistry.chemical_compound ,Adipocyte ,Adipocytes ,Glucose homeostasis ,Homeostasis ,Insulin ,Phosphorylation ,Cells, Cultured ,Adiposity ,Glucose Transporter Type 4 ,biology ,Forkhead Box Protein O1 ,Cell biology ,RNA Interference ,GRB2 ,Signal transduction ,Ion Channel Gating ,Signal Transduction ,medicine.medical_specialty ,Carbohydrate metabolism ,Transfection ,03 medical and health sciences ,Insulin resistance ,Chloride Channels ,Internal medicine ,medicine ,Animals ,Humans ,Obesity ,Cell Size ,GRB2 Adaptor Protein ,Glycogen Synthase Kinase 3 beta ,Membrane Proteins ,Cell Biology ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Glucose ,chemistry ,biology.protein ,Insulin Resistance ,Phosphatidylinositol 3-Kinase ,Energy Metabolism ,Proto-Oncogene Proteins c-akt - Abstract
Adipocytes undergo considerable volumetric expansion in the setting of obesity. It has been proposed that such marked increases in adipocyte size may be sensed via adipocyte-autonomous mechanisms to mediate size-dependent intracellular signalling. Here, we show that SWELL1 (LRRC8a), a member of the Leucine-Rich Repeat Containing protein family, is an essential component of a volume-sensitive ion channel (VRAC) in adipocytes. We find that SWELL1-mediated VRAC is augmented in hypertrophic murine and human adipocytes in the setting of obesity. SWELL1 regulates adipocyte insulin-PI3K-AKT2-GLUT4 signalling, glucose uptake and lipid content via SWELL1 C-terminal leucine-rich repeat domain interactions with GRB2/Cav1. Silencing GRB2 in SWELL1 KO adipocytes rescues insulin-pAKT2 signalling. In vivo, shRNA-mediated SWELL1 knockdown and adipose-targeted SWELL1 knockout reduce adiposity and adipocyte size in obese mice while impairing systemic glycaemia and insulin sensitivity. These studies identify SWELL1 as a cell-autonomous sensor of adipocyte size that regulates adipocyte growth, insulin sensitivity and glucose tolerance.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.