227 results on '"Chou-Long Huang"'
Search Results
2. WNK1 is required during male pachynema to sustain fertility
- Author
-
Ru-pin Alicia Chi, Xiaojiang Xu, Jian-Liang Li, Xin Xu, Guang Hu, Paula Brown, Cynthia Willson, Oleksandr Kirsanov, Christopher Geyer, Chou-Long Huang, Marcos Morgan, and Francesco DeMayo
- Subjects
Physiology ,Molecular biology ,Cell biology ,Omics ,Transcriptomics ,Science - Abstract
Summary: WNK1 is an important regulator in many physiological functions, yet its role in male reproduction is unexplored. In the male germline, WNK1 is upregulated in preleptotene spermatocytes indicating possible function(s) in spermatogenic meiosis. Indeed, deletion of Wnk1 in mid-pachytene spermatocytes using the Wnt7a-Cre mouse led to male sterility which resembled non-obstructive azoospermia in humans, where germ cells failed to complete spermatogenesis and produced no sperm. Mechanistically, we found elevated MTOR expression and signaling in the Wnk1-depleted spermatocytes. As MTOR is a central mediator of translation, we speculated that translation may be accelerated in these spermatocytes. Supporting this, we found the acrosome protein, ACRBP to be prematurely expressed in the spermatocytes with Wnk1 deletion. Our study uncovered an MTOR-regulating factor in the male germline with potential implications in translation, and future studies will aim to understand how WNK1 regulates MTOR activity and impact translation on a broader spectrum.
- Published
- 2023
- Full Text
- View/download PDF
3. WNK1 promotes water homeostasis by acting as a central osmolality sensor for arginine vasopressin release
- Author
-
Xin Jin, Jian Xie, Chia-Wei Yeh, Jen-Chi Chen, Chih-Jen Cheng, Cheng-Chang Lien, and Chou-Long Huang
- Subjects
Endocrinology ,Nephrology ,Medicine - Abstract
Maintaining internal osmolality constancy is essential for life. Release of arginine vasopressin (AVP) in response to hyperosmolality is critical. Current hypotheses for osmolality sensors in circumventricular organs (CVOs) of the brain focus on mechanosensitive membrane proteins. The present study demonstrated that intracellular protein kinase WNK1 was involved. Focusing on vascular-organ-of-lamina-terminalis (OVLT) nuclei, we showed that WNK1 kinase was activated by water restriction. Neuron-specific conditional KO (cKO) of Wnk1 caused polyuria with decreased urine osmolality that persisted in water restriction and blunted water restriction–induced AVP release. Wnk1 cKO also blunted mannitol-induced AVP release but had no effect on osmotic thirst response. The role of WNK1 in the osmosensory neurons in CVOs was supported by neuronal pathway tracing. Hyperosmolality-induced increases in action potential firing in OVLT neurons was blunted by Wnk1 deletion or pharmacological WNK inhibitors. Knockdown of Kv3.1 channel in OVLT by shRNA reproduced the phenotypes. Thus, WNK1 in osmosensory neurons in CVOs detects extracellular hypertonicity and mediates the increase in AVP release by activating Kv3.1 and increasing action potential firing from osmosensory neurons.
- Published
- 2023
- Full Text
- View/download PDF
4. Glucosylceramide synthase inhibition protects against cardiac hypertrophy in chronic kidney disease
- Author
-
Gabriel C. Baccam, Jian Xie, Xin Jin, Hyejung Park, Bing Wang, Hervé Husson, Oxana Ibraghimov-Beskrovnaya, and Chou-Long Huang
- Subjects
Medicine ,Science - Abstract
Abstract A significant population of patients with chronic kidney disease (CKD) develops cardiac hypertrophy, which can lead to heart failure and sudden cardiac death. Soluble klotho (sKL), the shed ectodomain of the transmembrane protein klotho, protects the heart against hypertrophic growth. We have shown that sKL protects the heart by regulating the formation and function of lipid rafts by targeting the sialic acid moiety of gangliosides, GM1/GM3. Reduction in circulating sKL contributes to an increased risk of cardiac hypertrophy in mice. sKL replacement therapy has been considered but its use is limited by the inability to mass produce the protein. Therefore, alternative methods to protect the heart are proposed. Glucosylation of ceramide catalyzed by glucosylceramide synthase is the entry step for the formation of gangliosides. Here we show that oral administration of a glucosylceramide synthase inhibitor (GCSi) reduces plasma and heart tissue glycosphingolipids, including gangliosides. Administration of GCSi is protective in two mouse models of cardiac stress-induction, one with isoproterenol overstimulation and the other with 5/6 nephrectomy-induced CKD. Treatment with GCSi does not alter the severity of renal dysfunction and hypertension in CKD. These results provide proof of principle for targeting glucosylceramide synthase to decrease gangliosides as a treatment for cardiac hypertrophy. They also support the hypothesis that sKL protects the heart by targeting gangliosides.
- Published
- 2022
- Full Text
- View/download PDF
5. CVVHD results in longer filter life than pre-filter CVVH: Results of a quasi-randomized clinical trial.
- Author
-
Lewis Mann, Patrick Ten Eyck, Chaorong Wu, Maria Story, Sree Jenigiri, Jayesh Patel, Iiro Honkanen, Kandi O'Connor, Janis Tener, Meenakshi Sambharia, Mony Fraer, Lama Nourredine, Douglas Somers, Jonathan Nizar, Lisa Antes, Sarat Kuppachi, Melissa Swee, Elizabeth Kuo, Chou-Long Huang, Diana I Jalal, and Benjamin R Griffin
- Subjects
Medicine ,Science - Abstract
BackgroundFilter clotting is a major issue in continuous kidney replacement therapy (CKRT) that interrupts treatment, reduces delivered effluent dose, and increases cost of care. While a number of variables are involved in filter life, treatment modality is an understudied factor. We hypothesized that filters in pre-filter continuous venovenous hemofiltration (CVVH) would have shorter lifespans than in continuous venovenous hemodialysis (CVVHD).MethodsThis was a single center, pragmatic, unblinded, quasi-randomized cluster trial conducted in critically ill adult patients with severe acute kidney injury (AKI) at the University of Iowa Hospitals and Clinics (UIHC) between March 2020 and December 2020. Patients were quasi-randomized by time block to receive pre-filter CVVH (convection) or CVVHD (diffusion). The primary outcome was filter life, and secondary outcomes were number of filters used, number of filters reaching 72 hours, and in-hospital mortality.ResultsIn the intention-to-treat analysis, filter life in pre-filter CVVH was 79% of that observed in CVVHD (mean ratio 0.79, 95% CI 0.65-0.97, p = 0.02). Median filter life (with interquartile range) in pre-filter CVVH was 21.8 (11.4-45.3) and was 26.6 (13.0-63.5) for CVVHD. In addition, 11.8% of filters in pre-filter CVVH were active for >72 hours, versus 21.2% in the CVVHD group. Finally, filter clotting accounted for the loss of 26.7% of filters in the CVVH group compared to 17.5% in the CVVHD group. There were no differences in overall numbers of filters used or mortality between groups.ConclusionsAmong critically patients with severe AKI requiring CKRT, use of pre-filter CVVH resulted in significantly shorter filter life compared to CVVHD.Trial registrationClinicalTrials.gov, NCT04762524. Registered 02/21/21-Retroactively registered, https://clinicaltrials.gov/ct2/show/NCT04762524?cond=The+Impact+of+CRRT+Modality+on+Filter+Life&draw=2&rank=1.
- Published
- 2023
- Full Text
- View/download PDF
6. Chloride sensing by WNK1 regulates NLRP3 inflammasome activation and pyroptosis
- Author
-
Lindsey Mayes-Hopfinger, Aura Enache, Jian Xie, Chou-Long Huang, Robert Köchl, Victor L. J. Tybulewicz, Teresa Fernandes-Alnemri, and Emad S. Alnemri
- Subjects
Science - Abstract
The serine/threonine kinase WNK1 is an inhibitor of chloride efflux. Here the authors show that this inhibition is a means of negatively regulating the activation of the NLRP3 inflammasome in macrophages, leading to reduced inflammatory responses.
- Published
- 2021
- Full Text
- View/download PDF
7. Impairment in renal medulla development underlies salt wasting in Clc-k2 channel deficiency
- Author
-
Meng-Hsuan Lin, Jen-Chi Chen, Xuejiao Tian, Chia-Ming Lee, I-Shing Yu, Yi-Fen Lo, Shinichi Uchida, Chou-Long Huang, Bi-Chang Chen, and Chih-Jen Cheng
- Subjects
Nephrology ,Medicine - Abstract
The prevailing view is that the ClC-Ka chloride channel (mouse Clc-k1) functions in the thin ascending limb to control urine concentration, whereas the ClC-Kb channel (mouse Clc-k2) functions in the thick ascending limb (TAL) to control salt reabsorption. Mutations of ClC-Kb cause classic Bartter syndrome, characterized by renal salt wasting, with perinatal to adolescent onset. We studied the roles of Clc-k channels in perinatal mouse kidneys using constitutive or inducible kidney-specific gene ablation and 2D and advanced 3D imaging of optically cleared kidneys. We show that Clc-k1 and Clc-k2 were broadly expressed and colocalized in perinatal kidneys. Deletion of Clc-k1 and Clc-k2 revealed that both participated in NKCC2- and NCC-mediated NaCl reabsorption in neonatal kidneys. Embryonic deletion of Clc-k2 caused tubular injury and impaired renal medulla and TAL development. Inducible deletion of Clc-k2 beginning after medulla maturation produced mild salt wasting resulting from reduced NCC activity. Thus, both Clc-k1 and Clc-k2 contributed to salt reabsorption in TAL and distal convoluted tubule (DCT) in neonates, potentially explaining the less-severe phenotypes in classic Bartter syndrome. As opposed to the current understanding that salt wasting in adult patients with Bartter syndrome is due to Clc-k2 deficiency in adult TAL, our results suggest that it originates mainly from defects occurring in the medulla and TAL during development.
- Published
- 2021
- Full Text
- View/download PDF
8. Loss of diacylglycerol kinase ε causes thrombotic microangiopathy by impairing endothelial VEGFA signaling
- Author
-
Dingxiao Liu, Qiong Ding, Dao-Fu Dai, Biswajit Padhy, Manasa K. Nayak, Can Li, Madison Purvis, Heng Jin, Chang Shu, Anil K. Chauhan, Chou-Long Huang, and Massimo Attanasio
- Subjects
Nephrology ,Medicine - Abstract
Loss of function of the lipid kinase diacylglycerol kinase ε (DGKε), encoded by the gene DGKE, causes a form of atypical hemolytic uremic syndrome that is not related to abnormalities of the alternative pathway of the complement, by mechanisms that are not understood. By generating a potentially novel endothelial specific Dgke-knockout mouse, we demonstrate that loss of Dgke in the endothelium results in impaired signaling downstream of VEGFR2 due to cellular shortage of phosphatidylinositol 4,5-biphosphate. Mechanistically, we found that, in the absence of DGKε in the endothelium, Akt fails to be activated upon VEGFR2 stimulation, resulting in defective induction of the enzyme cyclooxygenase 2 and production of prostaglandin E2 (PGE2). Treating the endothelial specific Dgke-knockout mice with a stable PGE2 analog was sufficient to reverse the clinical manifestations of thrombotic microangiopathy and proteinuria, possibly by suppressing the expression of matrix metalloproteinase 2 through PGE2-dependent upregulation of the chemokine receptor CXCR4. Our study reveals a complex array of autocrine signaling events downstream of VEGFR2 that are mediated by PGE2, that control endothelial activation and thrombogenic state, and that result in abnormalities of the glomerular filtration barrier.
- Published
- 2021
- Full Text
- View/download PDF
9. WNK1 regulates uterine homeostasis and its ability to support pregnancy
- Author
-
Ru-pin Alicia Chi, Tianyuan Wang, Chou-Long Huang, San-pin Wu, Steven L. Young, John P. Lydon, and Francesco J. DeMayo
- Subjects
Reproductive biology ,Medicine - Abstract
WNK1 (with no lysine [K] kinase 1) is an atypical kinase protein ubiquitously expressed in humans and mice. A mutation in its encoding gene causes hypertension in humans, which is associated with abnormal ion homeostasis. WNK1 is critical for in vitro decidualization in human endometrial stromal cells, thereby demonstrating its importance in female reproduction. Using a mouse model, WNK1 was ablated in the female reproductive tract to define its in vivo role in uterine biology. Loss of WNK1 altered uterine morphology, causing endometrial epithelial hyperplasia, adenomyotic features, and a delay in embryo implantation, ultimately resulting in compromised fertility. Combining transcriptomic, proteomic, and interactomic analyses revealed a potentially novel regulatory pathway whereby WNK1 represses AKT phosphorylation through protein phosphatase 2A (PP2A) in endometrial cells from both humans and mice. We show that WNK1 interacted with PPP2R1A, the alpha isoform of the PP2A scaffold subunit. This maintained the levels of PP2A subunits and stabilized its activity, which then dephosphorylated AKT. Therefore, loss of WNK1 reduced PP2A activity, causing AKT hypersignaling. Using FOXO1 as a readout of AKT activity, we demonstrate that there was escalated FOXO1 phosphorylation and nuclear exclusion, leading to a disruption in the expression of genes that are crucial for embryo implantation.
- Published
- 2020
- Full Text
- View/download PDF
10. Munc13 mediates klotho-inhibitable diacylglycerol-stimulated exocytotic insertion of pre-docked TRPC6 vesicles.
- Author
-
Jian Xie, Sung-Wan An, Xin Jin, Yuan Gui, and Chou-Long Huang
- Subjects
Medicine ,Science - Abstract
α-Klotho is a type 1 transmembrane protein that exhibits aging suppression function. The large amino-terminal extracellular domain of α-klotho is shed as soluble klotho (sKlotho) and functions as a circulating cardioprotective hormone. Diacylglycerol (DAG)-activated calcium-permeable TRPC6 channel plays a critical role in stress-induced cardiac remodeling. DAG activates TRPC6 by acting directly on the channel to increase its activity and by stimulation of channel exocytosis. sKlotho protects the heart by inhibiting DAG stimulation of TRPC6 exocytosis. How DAG stimulates TRPC6 exocytosis and thereby inhibition by sKlotho are unknown. Using a compound that directly activates TRPC6 without affecting channel exocytosis, we validate that sKlotho selectively blocks DAG stimulation of channel exocytosis. We further show that DAG stimulates exocytosis of TRPC6-containing vesicles pre-docked to the plasma membrane. Mnuc13 family proteins play important roles in the proper assembly of SNARE proteins and priming the vesicle competent for fusion. We show that DAG stimulates TRPC6 exocytosis by targeting to the C1 domain of Munc13-2. The results provide fresh insights into the molecular mechanism by which DAG regulates vesicle fusion and how sKlotho protects the heart against injury.
- Published
- 2020
- Full Text
- View/download PDF
11. New Insights into the Mechanism of Action of Soluble Klotho
- Author
-
George D. Dalton, Jian Xie, Sung-Wan An, and Chou-Long Huang
- Subjects
klotho ,FGF23 ,lipid rafts ,aging ,TRPC6 ,sialidase ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
The klotho gene encodes a type I single-pass transmembrane protein that contains a large extracellular domain, a membrane spanning segment, and a short intracellular domain. Klotho protein exists in several forms including the full-length membrane form (mKl) and a soluble circulating form [soluble klotho (sKl)]. mKl complexes with fibroblast growth factor receptors to form coreceptors for FGF23, which allows it to participate in FGF23-mediated signal transduction and regulation of phosphate and calcium homeostasis. sKl is present in the blood, urine, and cerebrospinal fluid where it performs a multitude of functions including regulation of ion channels/transporters and growth factor signaling. How sKl exerts these pleiotropic functions is poorly understood. One hurdle in understanding sKl’s mechanism of action as a “hormone” has been the inability to identify a receptor that mediates its effects. In the body, the kidneys are a major source of sKl and sKl levels decline during renal disease. sKl deficiency in chronic kidney disease makes the heart susceptible to stress-induced injury. Here, we summarize the current knowledge of mKl’s mechanism of action, the mechanistic basis of sKl’s protective, FGF23-independent effects on the heart, and provide new insights into the mechanism of action of sKl focusing on recent findings that sKl binds sialogangliosides in membrane lipid rafts to regulate growth factor signaling.
- Published
- 2017
- Full Text
- View/download PDF
12. Zebrafish WNK lysine deficient protein kinase 1 (wnk1) affects angiogenesis associated with VEGF signaling.
- Author
-
Ju-Geng Lai, Su-Mei Tsai, Hsiao-Chen Tu, Wen-Chuan Chen, Fong-Ji Kou, Jeng-Wei Lu, Horng-Dar Wang, Chou-Long Huang, and Chiou-Hwa Yuh
- Subjects
Medicine ,Science - Abstract
The WNK1 (WNK lysine deficient protein kinase 1) protein is a serine/threonine protein kinase with emerging roles in cancer. WNK1 causes hypertension and hyperkalemia when overexpressed and cardiovascular defects when ablated in mice. In this study, the role of Wnk1 in angiogenesis was explored using the zebrafish model. There are two zebrafish wnk1 isoforms, wnk1a and wnk1b, and both contain all the functional domains found in the human WNK1 protein. Both isoforms are expressed in the embryo at the initiation of angiogenesis and in the posterior cardinal vein (PCV), similar to fms-related tyrosine kinase 4 (flt4). Using morpholino antisense oligonucleotides against wnk1a and wnk1b, we observed that wnk1 morphants have defects in angiogenesis in the head and trunk, similar to flk1/vegfr2 morphants. Furthermore, both wnk1a and wnk1b mRNA can partially rescue the defects in vascular formation caused by flk1/vegfr2 knockdown. Mutation of the kinase domain or the Akt/PI3K phosphorylation site within wnk1 destroys this rescue capability. The rescue experiments provide evidence that wnk1 is a downstream target for Vegfr2 (vascular endothelial growth factor receptor-2) and Akt/PI3K signaling and thereby affects angiogenesis in zebrafish embryos. Furthermore, we found that knockdown of vascular endothelial growth factor receptor-2 (flk1/vegfr2) or vascular endothelial growth factor receptor-3 (flt4/vegfr3) results in a decrease in wnk1a expression, as assessed by in situ hybridization and q-RT-PCR analysis. Thus, the Vegf/Vegfr signaling pathway controls angiogenesis in zebrafish via Akt kinase-mediated phosphorylation and activation of Wnk1 as well as transcriptional regulation of wnk1 expression.
- Published
- 2014
- Full Text
- View/download PDF
13. Channel Function of Polycystin-2 in the Endoplasmic Reticulum Protects against Autosomal Dominant Polycystic Kidney Disease
- Author
-
Biswajit Padhy, Jian Xie, Runping Wang, Fang Lin, and Chou-Long Huang
- Subjects
Mice ,Potassium Channels ,TRPP Cation Channels ,Nephrology ,Potassium ,Animals ,Calcium ,General Medicine ,Endoplasmic Reticulum ,Polycystic Kidney, Autosomal Dominant ,Inositol ,Ion Channels ,Zebrafish - Abstract
Mutations ofThe trimeric intracellular cation (TRIC) channel TRIC-B is an ER-resident potassium channel that mediates potassium-calcium counterion exchange for inositol trisphosphate-mediated calcium ion release. Using TRIC-B as a tool, we examined the function of ER-localized polycystin-2 and its role in ADPKD pathogenesis in cultured cells, zebrafish, and mouse models.Agonist-induced ER calcium ion release was defective in cells lacking polycystin-2 and reversed by exogenous expression of TRIC-B.Polycystin-2 in the ER appears to be critical for anticystogenesis and likely functions as a potassium ion channel to facilitate potassium-calcium counterion exchange for inositol trisphosphate-mediated calcium release. The results advance the understanding of ADPKD pathogenesis and provides proof of principle for pharmacotherapy by TRIC-B activators.
- Published
- 2022
- Full Text
- View/download PDF
14. L-WNK1 is required for BK channel activation in intercalated cells
- Author
-
Allison L. Marciszyn, Tracey Lam, Lubika J. Nkashama, Daniel Flores, Rolando Carrisoza-Gaytán, Mohammad M. Al-bataineh, Shawn E. Griffiths, Arohan R. Subramanya, Jingxin Chen, Thomas R. Kleyman, Evan C. Ray, Aaliyah Winfrey, Chou Long Huang, Peng Wu, Wen-Hui Wang, and Lisa M. Satlin
- Subjects
0301 basic medicine ,Gene isoform ,BK channel ,Physiology ,030232 urology & nephrology ,Kidney ,Excretion ,Mice ,03 medical and health sciences ,0302 clinical medicine ,WNK Lysine-Deficient Protein Kinase 1 ,Animals ,Intercalated Cell ,Secretion ,Large-Conductance Calcium-Activated Potassium Channels ,Ion Transport ,biology ,Chemistry ,Kinase ,Nephrons ,WNK1 ,Molecular biology ,BK channel activity ,030104 developmental biology ,Potassium ,biology.protein ,Research Article - Abstract
Large-conductance K(+) (BK) channels expressed in intercalated cells (ICs) in the aldosterone-sensitive distal nephron (ASDN) mediate flow-induced K(+) secretion. In the ASDN of mice and rabbits, IC BK channel expression and activity increase with a high-K(+) diet. In cell culture, the long isoform of with-no-lysine kinase 1 (L-WNK1) increases BK channel expression and activity. Apical L-WNK1 expression is selectively enhanced in ICs in the ASDN of rabbits on a high-K(+) diet, suggesting that L-WNK1 contributes to BK channel regulation by dietary K(+). We examined the role of IC L-WNK1 expression in enhancing BK channel activity in response to a high-K(+) diet. Mice with IC-selective deletion of L-WNK1 (IC-L-WNK1-KO) and littermate control mice were placed on a high-K(+) (5% K(+), as KCl) diet for 10 or more days. IC-L-WNK1-KO mice exhibited reduced IC apical + subapical α-subunit expression and BK channel-dependent whole cell currents compared with controls. Six-hour urinary K(+) excretion in response a saline load was similar in IC-L-WNK1-KO mice and controls. The observations that IC-L-WNK1-KO mice on a high-K(+) diet have higher blood K(+) concentration and reduced IC BK channel activity are consistent with impaired urinary K(+) secretion, demonstrating that IC L-WNK1 has a role in the renal adaptation to a high-K(+) diet. NEW & NOTEWORTHY When mice are placed on a high-K(+) diet, genetic disruption of the long form of with no lysine kinase 1 (L-WNK1) in intercalated cells reduced relative apical + subapical localization of the large-conductance K(+) channel, blunted large-conductance K(+) channel currents in intercalated cells, and increased blood K(+) concentration. These data confirm an in vivo role of L-WNK1 in intercalated cells in adaptation to a high-K(+) diet.
- Published
- 2021
- Full Text
- View/download PDF
15. Chloride sensing by WNK1 regulates NLRP3 inflammasome activation and pyroptosis
- Author
-
Emad S. Alnemri, Teresa Fernandes-Alnemri, Jian Xie, Victor L. J. Tybulewicz, Aura Enache, Robert Köchl, Lindsey Mayes-Hopfinger, and Chou Long Huang
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,Pyrrolidines ,Inflammasomes ,Neutrophils ,Interleukin-1beta ,General Physics and Astronomy ,Inflammasome ,0302 clinical medicine ,WNK Lysine-Deficient Protein Kinase 1 ,Conditional gene knockout ,Multidisciplinary ,integumentary system ,Chemistry ,Caspase 1 ,Pyroptosis ,Imidazoles ,Cell biology ,030220 oncology & carcinogenesis ,Female ,Intracellular ,medicine.drug ,Cell death ,Science ,Protein Serine-Threonine Kinases ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,Chlorides ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Kinase activity ,Monocytes and macrophages ,Innate immune system ,L-Lactate Dehydrogenase ,Macrophages ,General Chemistry ,Immunity, Innate ,Mice, Inbred C57BL ,Tamoxifen ,030104 developmental biology ,Potassium - Abstract
The NLRP3 inflammasome mediates the production of proinflammatory cytokines and initiates inflammatory cell death. Although NLRP3 is essential for innate immunity, aberrant NLRP3 inflammasome activation contributes to a wide variety of inflammatory diseases. Understanding the pathways that control NLRP3 activation will help develop strategies to treat these diseases. Here we identify WNK1 as a negative regulator of the NLRP3 inflammasome. Macrophages deficient in WNK1 protein or kinase activity have increased NLRP3 activation and pyroptosis compared with control macrophages. Mice with conditional knockout of WNK1 in macrophages have increased IL-1β production in response to NLRP3 stimulation compared with control mice. Mechanistically, WNK1 tempers NLRP3 activation by balancing intracellular Cl– and K+ concentrations during NLRP3 activation. Collectively, this work shows that the WNK1 pathway has a critical function in suppressing NLRP3 activation and suggests that pharmacological inhibition of this pathway to treat hypertension might have negative clinical implications., The serine/threonine kinase WNK1 is an inhibitor of chloride efflux. Here the authors show that this inhibition is a means of negatively regulating the activation of the NLRP3 inflammasome in macrophages, leading to reduced inflammatory responses.
- Published
- 2021
16. WNK1 enforces macrophage lineage fidelity
- Author
-
Justin Perry, Alissa Trzeciak, Waleska Saitz Rojas, Adam Krebs, Giulia Zago, Ya-Ting Wang, Zhaoquan Wang, Pedro Saavedra, Jian Xie, Chou-Long Huang, Michael Overholtzer, Michael Glickman, Christopher Parkhurst, Thomas Vierbuchen, and Christopher Lucas
- Abstract
The appropriate development of myeloid progenitors into macrophages, the body’s professional phagocyte, is essential for organismal development, especially in mammals1. This dependence is exemplified by the observation that loss-of-function mutation in colony stimulating factor 1 receptor (CSF1R) results in multiple tissue abnormalities including osteopetrosis2. Despite this importance, little is known about the molecular and cell biological regulation of macrophage development. Here, we report the surprising finding that the chloride-sensing kinase With-no-lysine 1 (WNK1) is required for embryonic development of tissue-resident macrophages (TRMs). Myeloid-specific deletion of Wnk1 caused a dramatic loss of TRMs and subsequently disrupted organ development, induced systemic neutrophilia, and resulted in mortality between 3 and 4 weeks of age. Specifically, we observed that WNK1 absence stalled macrophage differentiation at the myeloid multipotent progenitor (MPP) stage, instead skewing MPP differentiation towards granulopoiesis. Mechanistically, the cognate CSF1R cytokine, macrophage-colony stimulating factor (M-CSF), triggers macropinocytosis in myeloid progenitors, which in turn induces phosphorylation of WNK1. Importantly, macropinocytosis by myeloid progenitors increases cytosolic chloride, which is directly sensed by WNK1. Perturbing chloride flux during macropinocytosis, inhibiting WNK1 chloride-sensing, and blocking macropinocytosis each skew progenitor differentiation from macrophage lineage to granulocyte lineage. Thus, we have uncovered a novel mechanism that links a cell biological process to a molecular circuit whereby WNK1 chloride-sensing and chloride flux act downstream of M-CSF-induced macropinocytosis by multipotent progenitors to ensure macrophage lineage fidelity.
- Published
- 2022
- Full Text
- View/download PDF
17. WNK1 Modulates mRNA Translation in Pachytene Spermatocytes and Is Indispensable for Male Fertility
- Author
-
Ru-pin Alicia Chi, Xiaojiang Xu, Jian-Liang Li, Xin Xu, Guang Hu, Paula Brown, Cynthia Willson, Oleksandr Kirsanov, Christopher B. Geyer, Chou-Long Huang, Marcos Morgan, and Francesco John DeMayo
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
- Full Text
- View/download PDF
18. LRRC8A reduces intracellular chloride to permit WNK activation in response to hypertonic stress
- Author
-
Elizabeth J. Goldsmith and Chou Long Huang
- Subjects
Multidisciplinary ,urogenital system ,Physiology ,Kinase ,Chemistry ,Protein subunit ,HEK 293 cells ,LRRC8A chloride channel ,Biological Sciences ,Cell biology ,NKCC ,Chlorides ,p38/MSK1 ,Osmotic Pressure ,Chloride channel ,RVI ,Phosphorylation ,osmostress ,Carrier Proteins ,Protein kinase A ,Cotransporter ,Intracellular - Abstract
Significance Rapid regulatory volume increase (RVI) is important for cell survival under hypertonic conditions. RVI is driven by Cl− uptake via the Na–K–Cl cotransporter (NKCC), which is activated by WNK kinases following a reduction in intracellular [Cl−]. However, how intracellular [Cl−] is regulated to modulate the WNK–NKCC axis and engage a protective RVI remains unknown. Our work reveals that LRRC8A-containing chloride channel is a key protective factor against hypertonic shocks. Considering that LRRC8A (SWELL1) is typically activated by low ionic strength under hypotonic stress, our results posed another interesting question: what activates this chloride channel under hypertonic stress? We demonstrated that, upon hyperosmotic activation, the p38-MSK1 pathway gates LRRC8A-containing chloride channel to facilitate activation of WNK–NKCC and an effective RVI., Regulation of cell volume is essential for tissue homeostasis and cell viability. In response to hypertonic stress, cells need rapid electrolyte influx to compensate water loss and to prevent cell death in a process known as regulatory volume increase (RVI). However, the molecular component able to trigger such a process was unknown to date. Using a genome-wide CRISPR/Cas9 screen, we identified LRRC8A, which encodes a chloride channel subunit, as the gene most associated with cell survival under hypertonic conditions. Hypertonicity activates the p38 stress-activated protein kinase pathway and its downstream MSK1 kinase, which phosphorylates and activates LRRC8A. LRRC8A-mediated Cl− efflux facilitates activation of the with-no-lysine (WNK) kinase pathway, which in turn, promotes electrolyte influx via Na+/K+/2Cl− cotransporter (NKCC) and RVI under hypertonic stress. LRRC8A-S217A mutation impairs channel activation by MSK1, resulting in reduced RVI and cell survival. In summary, LRRC8A is key to bidirectional osmotic stress responses and cell survival under hypertonic conditions.
- Published
- 2021
- Full Text
- View/download PDF
19. Soluble klotho regulates TRPC6 calcium signaling via lipid rafts, independent of the FGFR‐FGF23 pathway
- Author
-
Jon D. Wright, Jian Xie, Chou Long Huang, Sung Wan An, and Carmay Lim
- Subjects
0301 basic medicine ,Gene isoform ,Chemistry ,urologic and male genital diseases ,Biochemistry ,Cell biology ,TRPC6 ,03 medical and health sciences ,Transient receptor potential channel ,030104 developmental biology ,0302 clinical medicine ,Ectodomain ,Fibroblast growth factor receptor ,Genetics ,Binding site ,Molecular Biology ,Lipid raft ,030217 neurology & neurosurgery ,Biotechnology ,Calcium signaling - Abstract
Soluble klotho (sKlotho), the shed ectodomain of α-klotho, protects the heart by down-regulating transient receptor potential canonical isoform 6 (TRPC6)-mediated calcium signaling. Binding to α2-3-sialyllactose moiety of gangliosides in lipid rafts and inhibition of raft-dependent signaling underlies the mechanism. A recent 3-A X-ray structure of sKlotho in complex with fibroblast growth factor receptor (FGFR) and fibroblast growth factor 23 (FGF23) indicates that its β6α6 loop might block access to the proposed binding site for α2-3-sialyllactose. It was concluded that sKlotho only functions in complex with FGFR and FGF23 and that sKlotho's pleiotropic effects all depend on FGF23. Here, we report that sKlotho can inhibit TRPC6 channels expressed in cells lacking endogenous FGFRs. Structural modeling and molecular docking show that a repositioned β6α6 loop allows sKlotho to bind α2-3-sialyllactose. Molecular dynamic simulations further show the α2-3-sialyllactose-bound sKlotho complex to be stable. Domains mimicking sKlotho's sialic acid-recognizing activity inhibit TRPC6. The results strongly support the hypothesis that sKlotho can exert effects independent of FGF23 and FGFR.-Wright, J. D., An, S.-W., Xie, J., Lim, C., Huang, C.-L. Soluble klotho regulates TRPC6 calcium signaling via lipid rafts, independent of the FGFR-FGF23 pathway.
- Published
- 2019
- Full Text
- View/download PDF
20. Loss of diacylglycerol kinase ε causes thrombotic microangiopathy by impairing endothelial VEGFA signaling
- Author
-
Massimo Attanasio, Anil K. Chauhan, Heng Jin, Biswajit Padhy, Dao-Fu Dai, Qiong Ding, Madison Purvis, Chou Long Huang, Chang Shu, Can Li, Manasa K. Nayak, and Dingxiao Liu
- Subjects
Phosphatidylinositol 4,5-Diphosphate ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Diacylglycerol Kinase ,Receptors, CXCR4 ,Thrombotic microangiopathy ,Endothelium ,Dinoprostone ,Endothelial activation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Glomerular Filtration Barrier ,Atypical hemolytic uremic syndrome ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Autocrine signalling ,Protein kinase B ,Atypical Hemolytic Uremic Syndrome ,Diacylglycerol kinase ,Mice, Knockout ,Thrombotic Microangiopathies ,Chemistry ,Microcirculation ,General Medicine ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,Cell biology ,Autocrine Communication ,Vascular endothelial growth factor A ,030104 developmental biology ,medicine.anatomical_structure ,Cyclooxygenase 2 ,Nephrology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Inositol phosphates ,Medicine ,Matrix Metalloproteinase 2 ,Endothelium, Vascular ,Proto-Oncogene Proteins c-akt ,Research Article - Abstract
Loss of function of the lipid kinase diacylglycerol kinase ε (DGKε), encoded by the gene DGKE, causes a form of atypical hemolytic uremic syndrome that is not related to abnormalities of the alternative pathway of the complement, by mechanisms that are not understood. By generating a potentially novel endothelial specific Dgke-knockout mouse, we demonstrate that loss of Dgke in the endothelium results in impaired signaling downstream of VEGFR2 due to cellular shortage of phosphatidylinositol 4,5-biphosphate. Mechanistically, we found that, in the absence of DGKε in the endothelium, Akt fails to be activated upon VEGFR2 stimulation, resulting in defective induction of the enzyme cyclooxygenase 2 and production of prostaglandin E2 (PGE2). Treating the endothelial specific Dgke-knockout mice with a stable PGE2 analog was sufficient to reverse the clinical manifestations of thrombotic microangiopathy and proteinuria, possibly by suppressing the expression of matrix metalloproteinase 2 through PGE2-dependent upregulation of the chemokine receptor CXCR4. Our study reveals a complex array of autocrine signaling events downstream of VEGFR2 that are mediated by PGE2, that control endothelial activation and thrombogenic state, and that result in abnormalities of the glomerular filtration barrier.
- Published
- 2021
- Full Text
- View/download PDF
21. Kidney-Specific WNK1 Amplifies NCC Responsiveness to Potassium Imbalance
- Author
-
Sean D. Stocker, Daniel J. Shiwarski, Kate Querry, Lubika J. Nkashama, Sophia A. Knoell, Chou Long Huang, Rebecca T. Beacham, Allison L. Marciszyn, Arohan R. Subramanya, Cary R. Boyd-Shiwarski, and Shawn E. Griffiths
- Subjects
urogenital system ,Kinase ,Reabsorption ,Chemistry ,Nephron ,WNK1 ,Cell biology ,medicine.anatomical_structure ,parasitic diseases ,embryonic structures ,medicine ,Phosphorylation ,Potassium deficiency ,Distal convoluted tubule ,Signal transduction - Abstract
The distal convoluted tubule (DCT) NaCl cotransporter NCC is activated by phosphorylation, a process that is potassium-regulated and dependent on With-No-Lysine (WNK) kinases. KS-WNK1, a kidney-specific WNK1 isoform lacking the kinase domain, controls WNK signaling pathway localization in the DCT. Its role in NCC regulation, however, is unresolved: while early studies proposed that KS-WNK1 functions as an NCC inhibitor, recent work suggests that it activates NCC. To evaluate the role of KS-WNK1 on potassium-dependent NCC regulation, we studied KS-WNK1-KO mice across a wide range of plasma K+ (2.0-9.0 mmol/L), induced by dietary maneuvers and diuretic challenges. Potassium-deprived KS-WNK1-KO mice exhibited low WNK-dependent NCC phosphorylation compared to littermates, indicating that KS-WNK1 activates NCC during K+ deficiency. In contrast, relative NCC phosphorylation was high in potassium-loaded KS-WNK1-KO mice, consistent with KS-WNK1-mediated NCC inhibition during hyperkalemia. An integrated analysis revealed that KS-WNK1 expands the dynamic range of NCC responsiveness to potassium, steepening the linear inverse relationship between NCC phosphorylation and plasma [K+]. The effect of KS-WNK1 deletion was strongest in potassium-restricted females, as they developed exaggerated hypokalemia and thiazide insensitivity due to low NCC activity. Taken together, these findings indicate that KS-WNK1 is a potassium-responsive signaling amplifier that converts small changes in [K+] into large effects on NCC phosphorylation. This effect predominates in females during potassium deficiency, when high NCC activity is required to maintain salt reabsorption without exacerbating K+ losses. These observations define the role of KS-WNK1 in NCC regulation, and identify a novel mechanism that contributes to sexual dimorphism in the mammalian nephron.
- Published
- 2021
- Full Text
- View/download PDF
22. Contributors
- Author
-
Farah Al Sabie, Justine Bacchetta, Vincent Brandenburg, Ming Chang Hu, Chang Huei Chen, Lingfeng Chen, Erica L. Clinkenbeard, Michael J. Econs, Daniela Egli-Spichtig, Reinhold G. Erben, Sarah Erem, Carlos C. Faraco, Seiji Fukumoto, Regina Goetz, Alexander Grabner, Dieter Haffner, Mark R. Hanudel, Ping He, Gunnar H. Heine, Martin Hewison, Jan-Luuk Hillebrands, Keith A. Hruska, Chou-Long Huang, Erik A. Imel, Anna Jovanovich, Stefanie Krick, Makoto Kuro-o, Seong Min Lee, Maren Leifheit-Nestler, Rik Mencke, Orson W. Moe, Moosa Mohammadi, John Musgrove, Javier A. Neyra, Hannes Olauson, Katherine Wesseling Perry, Farzana Perwad, J. Wesley Pike, Mohammed S. Razzaque, Beatrice Richter, Isidro B. Salusky, Renal Section, Sarah Seiler-Mußler, Taylor Struemph, Marc G. Vervloet, Curtis Vrabec, Matthew J. Williams, Clinton B. Wright, and Allen Zinkle
- Published
- 2021
- Full Text
- View/download PDF
23. Soluble α-klotho—a fibroblast growth factor 23–independent hormone
- Author
-
Orson W. Moe and Chou Long Huang
- Subjects
Paracrine signalling ,Ectodomain ,Fibroblast growth factor receptor ,Chemistry ,Cell surface receptor ,Lipid microdomain ,lipids (amino acids, peptides, and proteins) ,Receptor ,Autocrine signalling ,Lipid raft ,Cell biology - Abstract
Membranous α-Klotho (mKlotho) interacts with fibroblast growth factor receptor (FGFR) to form coreceptors for fibroblast growth factor 23 (FGF23). The ectodomain of mKlotho is shed (soluble Klotho; sKlotho) and functions as a circulating endocrine or local autocrine/paracrine factor. sKlotho exerts pleiotropic actions. Identification of membrane receptor(s) is essential for designating sKlotho to have “hormonal” function. Lipid rafts are membrane microdomain important in many cellular processes. Here, we review recent studies reporting that sialogangliosides of membrane lipid rafts serve as receptors for sKlotho. The X-ray crystal structure of sKlotho in ternary complex with FGFR and FGF23 has been solved. However, sKlotho binding to lipid rafts does not require FGF23 or FGFR. Computational studies including modeling, docking, and molecular dynamic simulations reveal that slight movement in the KL1 domain of sKlotho allows stable binding of the sialyllactose, headgroup of sialogangliosides. Thus, sKlotho satisfies the criteria to be an FGF23-independent hormone. Binding to lipid rafts may underlie the pleiotropic actions of sKlotho that seem to permeate all cells.
- Published
- 2021
- Full Text
- View/download PDF
24. WNK1 regulates uterine homeostasis and its ability to support pregnancy
- Author
-
Steven L. Young, Tianyuan Wang, Ru Pin Alicia Chi, Francesco J. DeMayo, Chou-long Huang, San-Pin Wu, and John P. Lydon
- Subjects
Phosphoprotein phosphatases ,Male ,0301 basic medicine ,Stromal cell ,Proteome ,Reproductive biology ,FOXO1 ,Obstetrics/gynecology ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,WNK Lysine-Deficient Protein Kinase 1 ,Pregnancy ,Animals ,Homeostasis ,Humans ,Gene Regulatory Networks ,Protein Phosphatase 2 ,Phosphorylation ,Protein kinase B ,Mice, Knockout ,Forkhead Box Protein O1 ,Reproduction ,Uterus ,Decidualization ,General Medicine ,Protein phosphatase 2 ,WNK1 ,Cell biology ,Mice, Inbred C57BL ,Fertility ,030104 developmental biology ,Ion homeostasis ,030220 oncology & carcinogenesis ,Medicine ,Female ,Transcriptome ,Proto-Oncogene Proteins c-akt ,Research Article - Abstract
WNK1 (with no lysine [K] kinase 1) is an atypical kinase protein ubiquitously expressed in humans and mice. A mutation in its encoding gene causes hypertension in humans, which is associated with abnormal ion homeostasis. WNK1 is critical for in vitro decidualization in human endometrial stromal cells, thereby demonstrating its importance in female reproduction. Using a mouse model, WNK1 was ablated in the female reproductive tract to define its in vivo role in uterine biology. Loss of WNK1 altered uterine morphology, causing endometrial epithelial hyperplasia, adenomyotic features, and a delay in embryo implantation, ultimately resulting in compromised fertility. Combining transcriptomic, proteomic, and interactomic analyses revealed a potentially novel regulatory pathway whereby WNK1 represses AKT phosphorylation through protein phosphatase 2A (PP2A) in endometrial cells from both humans and mice. We show that WNK1 interacted with PPP2R1A, the alpha isoform of the PP2A scaffold subunit. This maintained the levels of PP2A subunits and stabilized its activity, which then dephosphorylated AKT. Therefore, loss of WNK1 reduced PP2A activity, causing AKT hypersignaling. Using FOXO1 as a readout of AKT activity, we demonstrate that there was escalated FOXO1 phosphorylation and nuclear exclusion, leading to a disruption in the expression of genes that are crucial for embryo implantation., The atypical kinase WNK1 is critical for maintaining uterine homeostasis and mediating proper implantation through the PP2A/AKT signaling pathway.
- Published
- 2020
25. Munc13 mediates klotho-inhibitable diacylglycerol-stimulated exocytotic insertion of pre-docked TRPC6 vesicles
- Author
-
Chou Long Huang, Xin Jin, Jian Xie, Yuan Gui, and Sung Wan An
- Subjects
0301 basic medicine ,Cell Membranes ,Toxicology ,Pathology and Laboratory Medicine ,Membrane Fusion ,TRPC6 ,0302 clinical medicine ,Medicine and Health Sciences ,Toxins ,Klotho ,Glucuronidase ,C1 domain ,Secretory Pathway ,Multidisciplinary ,Chemistry ,Cell biology ,Laboratory Equipment ,Cell Processes ,Engineering and Technology ,Cell lines ,Medicine ,lipids (amino acids, peptides, and proteins) ,Cellular Structures and Organelles ,Biological cultures ,Research Article ,Vesicle fusion ,Science ,Toxic Agents ,Equipment ,Nerve Tissue Proteins ,Exocytosis ,Diglycerides ,03 medical and health sciences ,TRPC6 Cation Channel ,Humans ,Vesicles ,Klotho Proteins ,Diacylglycerol kinase ,Pipettes ,urogenital system ,HEK 293 cells ,Cell Membrane ,Biology and Life Sciences ,Membrane Proteins ,Lipid bilayer fusion ,Cell Biology ,Research and analysis methods ,HEK293 Cells ,030104 developmental biology ,Membrane protein ,030217 neurology & neurosurgery - Abstract
α-Klotho is a type 1 transmembrane protein that exhibits aging suppression function. The large amino-terminal extracellular domain of α-klotho is shed as soluble klotho (sKlotho) and functions as a circulating cardioprotective hormone. Diacylglycerol (DAG)-activated calcium-permeable TRPC6 channel plays a critical role in stress-induced cardiac remodeling. DAG activates TRPC6 by acting directly on the channel to increase its activity and by stimulation of channel exocytosis. sKlotho protects the heart by inhibiting DAG stimulation of TRPC6 exocytosis. How DAG stimulates TRPC6 exocytosis and thereby inhibition by sKlotho are unknown. Using a compound that directly activates TRPC6 without affecting channel exocytosis, we validate that sKlotho selectively blocks DAG stimulation of channel exocytosis. We further show that DAG stimulates exocytosis of TRPC6-containing vesicles pre-docked to the plasma membrane. Mnuc13 family proteins play important roles in the proper assembly of SNARE proteins and priming the vesicle competent for fusion. We show that DAG stimulates TRPC6 exocytosis by targeting to the C1 domain of Munc13-2. The results provide fresh insights into the molecular mechanism by which DAG regulates vesicle fusion and how sKlotho protects the heart against injury.
- Published
- 2020
26. Chloride oscillation in pacemaker neurons regulates circadian rhythms through a chloride-sensing WNK kinase signaling cascade
- Author
-
Chou Long Huang, Adrian Rothenfluh, Sung-Wan An, Gaelle Mercenne, Jeffrey N. Schellinger, Iris Titos, John M. Pleinis, Jianrui Hu, Aylin R. Rodan, and Qifei Sun
- Subjects
Mammals ,Neurons ,GABAA receptor ,Chemistry ,Period (gene) ,Protein Serine-Threonine Kinases ,Chloride ,Potassium channel ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Circadian Rhythm ,medicine.anatomical_structure ,Drosophila melanogaster ,Chlorides ,Chloride channel ,medicine ,Animals ,Drosophila Proteins ,Neuron ,Circadian rhythm ,General Agricultural and Biological Sciences ,Intracellular ,medicine.drug ,Signal Transduction - Abstract
Central pacemaker neurons regulate circadian rhythms and undergo diurnal variation in electrical activity in mammals and flies. In mammals, circadian variation in the intracellular chloride concentration of pacemaker neurons has been proposed to influence the response to GABAergic neurotransmission through GABAA receptor chloride channels. However, results have been contradictory, and a recent study demonstrated circadian variation in pacemaker neuron chloride without an effect on GABA response. Therefore, whether and how intracellular chloride regulates circadian rhythms remains controversial. Here, we demonstrate a signaling role for intracellular chloride in the Drosophila ventral lateral (LNv) pacemaker neurons. In control flies, intracellular chloride increases in LNv neurons over the course of the morning. Chloride transport through the sodium-potassium-2-chloride (NKCC) and potassium-chloride (KCC) cotransporters is a major determinant of intracellular chloride concentrations. Drosophila melanogaster with loss-of-function mutations in the NKCC encoded by Ncc69 have abnormally low intracellular chloride six hours after lights on, and a lengthened circadian period. Loss of kcc, which is expected to increase intracellular chloride, suppresses the long-period phenotype of Ncc69 mutant flies. Activation of a chloride-inhibited kinase cascade, consisting of the WNK (With No Lysine (K)) kinase and its downstream substrate, Fray, is necessary and sufficient to prolong period length. Fray activation of an inwardly rectifying potassium channel, Irk1, is also required for the long-period phenotype. These results indicate that the NKCC-dependent rise in intracellular chloride in Drosophila LNv pacemaker neurons restrains WNK-Fray signaling and overactivation of an inwardly rectifying potassium channel to maintain normal circadian period length.
- Published
- 2022
- Full Text
- View/download PDF
27. Differential roles of WNK4 in regulation of NCC in vivo
- Author
-
Yih Sheng Yang, Sung Sen Yang, Chou Long Huang, Jian Xie, and Shih-Hua Lin
- Subjects
0301 basic medicine ,Physiology ,Injections, Subcutaneous ,Sodium Chloride Symporter Inhibitors ,030232 urology & nephrology ,Protein Serine-Threonine Kinases ,Sodium Chloride ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,0302 clinical medicine ,parasitic diseases ,medicine ,Animals ,Solute Carrier Family 12, Member 3 ,Distal convoluted tubule ,Phosphorylation ,Kidney Tubules, Distal ,Mice, Knockout ,urogenital system ,Reabsorption ,Chemistry ,Wild type ,Potassium, Dietary ,Biological Transport ,WNK1 ,Renal Reabsorption ,Cell biology ,WNK4 ,Mice, Inbred C57BL ,Renal Elimination ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,Cotransporter ,Intracellular ,Research Article - Abstract
The Na+-Cl− cotransporter (NCC) in distal convoluted tubule (DCT) plays important roles in renal NaCl reabsorption. The current hypothesis for the mechanism of regulation of NCC focuses on WNK4 and intracellular Cl− concentration ([Cl−]i). WNK kinases bind Cl−, and Cl− binding decreases the catalytic activity. It is believed that hypokalemia under low K+ intake decreases [Cl−]i to activate WNK4, which thereby phosphorylates and stimulates NCC through activation of SPAK. However, increased NCC activity and apical NaCl entry would mitigate the fall in [Cl−]i. Whether [Cl−]i in DCT under low-K+ diet is sufficiently low to activate WNK4 is unknown. Furthermore, increased luminal NaCl delivery also stimulates NCC and causes upregulation of the transporter. Unlike low K+ intake, increased luminal NaCl delivery would tend to increase [Cl−]i. Thus we investigated the role of WNK4 and [Cl−]i in regulating NCC. We generated Wnk4-knockout mice and examined regulation of NCC by low K+ intake and by increased luminal NaCl delivery in knockout (KO) and wild-type mice. Wnk4-KO mice have marked reduction in the abundance, phosphorylation, and functional activity of NCC vs. wild type. Low K+ intake increases NCC phosphorylation and functional activity in wild-type mice, but not in Wnk4-KO mice. Increased luminal NaCl delivery similarly upregulates NCC, which, contrary to low K+ intake, is not abolished in Wnk4-KO mice. The results reveal that modulation of WNK4 activity by [Cl−]i is not the sole mechanism for regulating NCC. Increased luminal NaCl delivery upregulates NCC via yet unknown mechanism(s) that may override inhibition of WNK4 by high [Cl−]i.
- Published
- 2018
- Full Text
- View/download PDF
28. OSR1 regulates a subset of inward rectifier potassium channels via a binding motif variant
- Author
-
Jonathan Z. Yang, Svetlana Earnest, Ashesh T. Trivedi, Steve Stippec, Sachith Gallolu Kankanamalage, Hamid Mirzaei, Melanie H. Cobb, Sung Wan An, Clinton A. Taylor, and Chou Long Huang
- Subjects
0301 basic medicine ,Amino Acid Motifs ,Molecular Sequence Data ,Protein domain ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Humans ,Amino Acid Sequence ,Potassium Channels, Inwardly Rectifying ,Ion transporter ,Multidisciplinary ,urogenital system ,Inward-rectifier potassium ion channel ,Chemistry ,Kinase ,Biological Sciences ,WNK1 ,Potassium channel ,Cell biology ,030104 developmental biology ,Multigene Family ,Mutation ,cardiovascular system ,Signal transduction ,Sequence motif ,Sequence Alignment ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
The with-no-lysine (K) (WNK) signaling pathway to STE20/SPS1-related proline- and alanine-rich kinase (SPAK) and oxidative stress-responsive 1 (OSR1) kinase is an important mediator of cell volume and ion transport. SPAK and OSR1 associate with upstream kinases WNK 1-4, substrates, and other proteins through their C-terminal domains which interact with linear R-F-x-V/I sequence motifs. In this study we find that SPAK and OSR1 also interact with similar affinity with a motif variant, R-x-F-x-V/I. Eight of 16 human inward rectifier K+ channels have an R-x-F-x-V motif. We demonstrate that two of these channels, Kir2.1 and Kir2.3, are activated by OSR1, while Kir4.1, which does not contain the motif, is not sensitive to changes in OSR1 or WNK activity. Mutation of the motif prevents activation of Kir2.3 by OSR1. Both siRNA knockdown of OSR1 and chemical inhibition of WNK activity disrupt NaCl-induced plasma membrane localization of Kir2.3. Our results suggest a mechanism by which WNK-OSR1 enhance Kir2.1 and Kir2.3 channel activity by increasing their plasma membrane localization. Regulation of members of the inward rectifier K+ channel family adds functional and mechanistic insight into the physiological impact of the WNK pathway.
- Published
- 2018
- Full Text
- View/download PDF
29. Emerging Targets of Diuretic Therapy
- Author
-
Aylin R. Rodan, Chou Long Huang, and Chih-Jen Cheng
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Urea transporter ,medicine.medical_treatment ,030232 urology & nephrology ,Pharmacology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Aquaretic ,Internal medicine ,Edema ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Diuretics ,Heart Failure ,biology ,Chemistry ,Reabsorption ,Pendrin ,medicine.disease ,High-Throughput Screening Assays ,030104 developmental biology ,Endocrinology ,Drug Design ,Heart failure ,Hypertension ,ROMK ,biology.protein ,medicine.symptom ,Diuretic - Abstract
Diuretics are commonly prescribed for treatment in patients with hypertension, edema or heart failure. Studies on hypertensive and salt-losing disorders and on urea transporters have contributed to better understanding of mechanisms of renal salt and water reabsorption and their regulation. Proteins involved in the regulatory pathways are emerging targets for diuretic and aquaretic therapy. Integrative high-throughput screening, protein structure analysis, and chemical modification have identified promising agents for pre-clinical testing in animals. These include WNK-SPAK inhibitors, ClC-K channel antagonists, ROMK channel antagonists, and pendrin and urea transporter inhibitors. We discuss the potential advantages and side effects of these potential diuretics.
- Published
- 2017
- Full Text
- View/download PDF
30. Functional severity ofCLCNKBmutations correlates with phenotypes in patients with classic Bartter's syndrome
- Author
-
Chou Long Huang, Jen Chi Chen, Yi Fen Lo, Shih-Hua Lin, and Chih-Jen Cheng
- Subjects
0301 basic medicine ,Genetics ,Mutation ,CLCNKB ,biology ,Physiology ,HEK 293 cells ,Mutant ,030232 urology & nephrology ,medicine.disease_cause ,Phenotype ,Molecular biology ,03 medical and health sciences ,Bartter's syndrome ,030104 developmental biology ,0302 clinical medicine ,medicine ,biology.protein ,Missense mutation ,Heterologous expression - Abstract
Mutations in CLCNKB gene encoding human voltage-gated chloride ClC-Kb (hClC-Kb) channel cause classic Bartter's syndrome (BS). In contrast to antenatal BS, classic BS manifests highly variable phenotypes. The functional severity of mutant channel has been proposed to explain this phenomenon. Due to difficulties in the expression of hClC-Kb in heterologous expression systems, the functional consequences of mutant channels haven't been thoroughly examined, and the genotype-phenotype association hasn't been established. In this study, we found that hClC-Kb, when expressed in human embryonic kidney (HEK) cells, was unstable due to degradation by proteasome. In-frame fusion of green fluorescent protein (GFP) to the C-terminus of the channel may ameliorate proteasome degradation. Co-expression of barttin increased protein abundance and membrane trafficking of hClC-Kb and markedly increased functional chloride current. We then functionally characterized eighteen missense mutations identified in our classic BS cohort and others using HEK cells expressing hClC-Kb-GFP. Most CLCNKB mutations resulted in marked reduction in protein abundance and chloride current, especially those residing at barttin-binding sites, dimer interface, and selectivity filter. We enrolled classic BS patients carrying homozygous missense mutations with well-described functional consequences and clinical presentations for genotype-phenotype analysis. We found significant correlations of mutant chloride current with the age at diagnosis, plasma chloride concentration, and urine calcium excretion rate. In conclusion, hClC-Kb expression in HEK cells is susceptible to proteasome degradation, and fusion of GFP to the C-terminus of hClC-Kb improves the protein expression. The functional severity of CLCNKB mutation is an important determinant of the phenotype in classic BS. This article is protected by copyright. All rights reserved
- Published
- 2017
- Full Text
- View/download PDF
31. WNK4 kinase is a physiological intracellular chloride sensor
- Author
-
Yi Fen Lo, Chih-Jen Cheng, Shih-Hua Lin, Ya Wen Lin, Jen Chi Chen, and Chou Long Huang
- Subjects
Multidisciplinary ,Chemistry ,urogenital system ,Potassium ,Pseudohypoaldosteronism ,chemistry.chemical_element ,Biological Sciences ,WNK1 ,medicine.disease ,WNK4 ,Dietary Potassium ,Cell biology ,medicine ,Cotransporter ,Homeostasis ,Intracellular - Abstract
With-no-lysine (WNK) kinases regulate renal sodium-chloride cotransporter (NCC) to maintain body sodium and potassium homeostasis. Gain-of-function mutations of WNK1 and WNK4 in humans lead to a Mendelian hypertensive and hyperkalemic disease pseudohypoaldosteronism type II (PHAII). X-ray crystal structure and in vitro studies reveal chloride ion (Cl(−)) binds to a hydrophobic pocket within the kinase domain of WNKs to inhibit its activity. The mechanism is thought to be important for physiological regulation of NCC by extracellular potassium. To test the hypothesis that WNK4 senses the intracellular concentration of Cl(−) physiologically, we generated knockin mice carrying Cl(−)-insensitive mutant WNK4. These mice displayed hypertension, hyperkalemia, hyperactive NCC, and other features fully recapitulating human and mouse models of PHAII caused by gain-of-function WNK4. Lowering plasma potassium levels by dietary potassium restriction increased NCC activity in wild-type, but not in knockin, mice. NCC activity in knockin mice can be further enhanced by the administration of norepinephrine, a known activator of NCC. Raising plasma potassium by oral gavage of potassium inactivated NCC within 1 hour in wild-type mice, but had no effect in knockin mice. The results provide compelling support for the notion that WNK4 is a bona fide physiological intracellular Cl(−) sensor and that Cl(−) regulation of WNK4 underlies the mechanism of regulation of NCC by extracellular potassium.
- Published
- 2019
32. WNK1 kinase balances T cell adhesion versus migration in vivo
- Author
-
Chou Long Huang, Lesley Vanes, Robert Köchl, Tiago F. Brazão, Flavian Thelen, Jens V. Stein, Kathryn Fountain, Victor L. J. Tybulewicz, Jian Xie, and Ruth Lyck
- Subjects
EXPRESSION ,0301 basic medicine ,PROTEIN-KINASES ,T-Lymphocytes ,Immunology ,Integrin ,Receptors, Lymphocyte Homing ,BLOOD-PRESSURE ,Protein Serine-Threonine Kinases ,Biology ,Kidney ,Article ,LEUKOCYTE MIGRATION ,Minor Histocompatibility Antigens ,Mice ,03 medical and health sciences ,WNK Lysine-Deficient Protein Kinase 1 ,Cell Movement ,Cell Adhesion ,Animals ,Homeostasis ,Solute Carrier Family 12, Member 2 ,Immunology and Allergy ,IL-2 receptor ,610 Medicine & health ,Cell adhesion ,Cells, Cultured ,Mice, Knockout ,Science & Technology ,ROLES ,Ion Transport ,INTEGRIN ACTIVATION ,Cell adhesion molecule ,Kidney metabolism ,Adhesion ,Protein-Serine-Threonine Kinases ,Cell biology ,Mice, Inbred C57BL ,COTRANSPORTER ,030104 developmental biology ,1107 Immunology ,T cell migration ,biology.protein ,RNA Interference ,Neural cell adhesion molecule ,REGULATOR ,Life Sciences & Biomedicine ,LFA-1 - Abstract
Adhesion and migration of T cells are controlled by chemokines and by adhesion molecules, especially integrins, and have critical roles in the normal physiological function of T lymphocytes. Using an RNA-mediated interference screen, we identified the WNK1 kinase as a regulator of both integrin-mediated adhesion and T cell migration. We found that WNK1 is a negative regulator of integrin-mediated adhesion, whereas it acts as a positive regulator of migration via the kinases OXSR1 and STK39 and the ion co-transporter SLC12A2. WNK1-deficient T cells home less efficiently to lymphoid organs and migrate more slowly through them. Our results reveal that a pathway previously known only to regulate salt homeostasis in the kidney functions to balance T cell adhesion and migration.
- Published
- 2016
- Full Text
- View/download PDF
33. L-WNK1 is required for BK channel activation in intercalated cells.
- Author
-
Ray, Evan C., Carrisoza-Gaytan, Rolando, Al-Bataineh, Mohammad, Marciszyn, Allison L., Nkashama, Lubika J., Jingxin Chen, Winfrey, Aaliyah, Griffiths, Shawn, Lam, Tracey R., Flores, Daniel, Peng Wu, WenHui Wang, Chou-Long Huang, Subramanya, Arohan R., Kleyman, Thomas R., and Satlin, Lisa M.
- Subjects
CELL culture ,KIDNEY tubules ,POTASSIUM channels ,RABBITS ,ANIMAL nutrition - Abstract
Large-conductance K
+ (BK) channels expressed in intercalated cells (ICs) in the aldosterone-sensitive distal nephron (ASDN) mediate flow-induced K+ secretion. In the ASDN of mice and rabbits, IC BK channel expression and activity increase with a high-K+ diet. In cell culture, the long isoform of with-no-lysine kinase 1 (L-WNK1) increases BK channel expression and activity. Apical L-WNK1 expression is selectively enhanced in ICs in the ASDN of rabbits on a high-K+ diet, suggesting that L-WNK1 contributes to BK channel regulation by dietary K+ . We examined the role of IC L-WNK1 expression in enhancing BK channel activity in response to a high-K+ diet. Mice with IC-selective deletion of L-WNK1 (IC-L-WNK1-KO) and littermate control mice were placed on a high-K+ (5% K+ , as KCl) diet for 10 or more days. IC-L-WNK1-KO mice exhibited reduced IC apical þ subapical α-subunit expression and BK channel-dependent whole cell currents compared with controls. Six-hour urinary K+ excretion in response a saline load was similar in IC-L-WNK1-KO mice and controls. The observations that IC-L-WNK1-KO mice on a high-K+ diet have higher blood K+ concentration and reduced IC BK channel activity are consistent with impaired urinary K+ secretion, demonstrating that IC L-WNK1 has a role in the renal adaptation to a high-K+ diet. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
34. WNK1 Kinase Stimulates Angiogenesis to Promote Tumor Growth and Metastasis
- Author
-
Horng-Dar Wang, Chiou-Hwa Yuh, Zong Lin Sie, Shu Chen Liu, Bonifasius Putera Sampurna, Chou Long Huang, Po Jui Hsu, and Ruei Yang Li
- Subjects
Cancer Research ,biology ,Chemistry ,Kinase ,Angiogenesis ,colorectal cancer ,hepatocellular carcinoma ,zebrafish ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease_cause ,WNK1 ,biology.organism_classification ,lcsh:RC254-282 ,Article ,tumor-induced angiogenesis ,WNK4 ,Vascular endothelial growth factor ,chemistry.chemical_compound ,Oncology ,medicine ,Cancer research ,Tumor promotion ,with-no-lysine (k) kinase i ,Carcinogenesis ,Zebrafish - Abstract
With-no-lysine (K)-1 (WNK1) is the founding member of family of four protein kinases with atypical placement of catalytic lysine that play important roles in regulating epithelial ion transport. Gain-of-function mutations of WNK1 and WNK4 cause a mendelian hypertension and hyperkalemic disease. WNK1 is ubiquitously expressed and essential for embryonic angiogenesis in mice. Increasing evidence indicates the role of WNK kinases in tumorigenesis at least partly by stimulating tumor cell proliferation. Here, we show that human hepatoma cells xenotransplanted into zebrafish produced high levels of vascular endothelial growth factor (VEGF) and WNK1, and induced expression of zebrafish wnk1. Knockdown of wnk1 in zebrafish decreased tumor-induced ectopic vessel formation and inhibited tumor proliferation. Inhibition of WNK1 or its downstream kinases OSR1 (oxidative stress responsive kinase 1)/SPAK (Ste20-related proline alanine rich kinase) using chemical inhibitors decreased ectopic vessel formation as well as proliferation of xenotransplanted hepatoma cells. The effect of WNK and OSR1 inhibitors is greater than that achieved by inhibitor of VEGF signaling cascade. These inhibitors also effectively inhibited tumorigenesis in two separate transgenic zebrafish models of intestinal and hepatocellular carcinomas. Endothelial-specific overexpression of wnk1 enhanced tumorigenesis in transgenic carcinogenic fish, supporting endothelial cell-autonomous effect of WNK1 in tumor promotion. Thus, WNK1 can promote tumorigenesis by multiple effects that include stimulating tumor angiogenesis. Inhibition of WNK1 may be a potent anti-cancer therapy.
- Published
- 2020
- Full Text
- View/download PDF
35. Protein‐Protein Interaction Prediction Yields WNK‐SPAK/OSR1 Pathway as Regulator of Kir2.1 and Kir2.3 K + Channels
- Author
-
Sachith Gallolu Kankanamalage, Clinton A. Taylor, Melanie Houston Cobb, Sung‐Wan An, and Chou Long Huang
- Subjects
Chemistry ,Genetics ,Kir2.1 ,Regulator ,Protein–protein interaction prediction ,Molecular Biology ,Biochemistry ,Biotechnology ,K channels ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
36. KS‐WNK1 Expands the Dynamic Range of NCC Regulation by Dietary Potassium
- Author
-
Kelly A. Connolly, Lubika J. Nkashama, Chou Long Huang, Cary R. Boyd-Shiwarski, and Arohan R. Subramanya
- Subjects
Chemistry ,Genetics ,Food science ,WNK1 ,Molecular Biology ,Biochemistry ,Biotechnology ,Dietary Potassium - Published
- 2018
- Full Text
- View/download PDF
37. Reply to Farfel et al.: Is enhanced chloride reabsorption in proximal tubule a possible mechanism of metabolic acidosis in PHAII?
- Author
-
Shih-Hua Lin, Chou Long Huang, Chih-Jen Cheng, and Jen Chi Chen
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Multidisciplinary ,Hyperkalemia ,urogenital system ,Reabsorption ,Sodium ,030232 urology & nephrology ,Pseudohypoaldosteronism ,chemistry.chemical_element ,Metabolic acidosis ,medicine.disease ,WNK4 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Internal medicine ,medicine ,Distal convoluted tubule ,medicine.symptom ,Kinase activity - Abstract
Hyperchloremic metabolic acidosis along with hypertension and hyperkalemia are features of pseudohypoaldosteronism type II (PHAII). Increased activity of sodium chloride cotransporter (NCC) is believed to be an important mechanism of these phenotypic features (1). Gain-of-function mutations of WNK4 in PHAII activate NCC in the distal convoluted tubule, which leads to enhanced sodium and chloride reabsorption causing hypertension and diminished sodium delivery to the downstream cortical collecting duct, resulting in hyperkalemia. Our recent report (2) that chloride-insensitive WNK4 knockin mouse exhibits increased WNK4 kinase activity and fully recapitulates PHAII phenotype supports the notion. Still, as mentioned by Farfel et al. (3), there remain debates on the … [↵][1]1To whom correspondence may be addressed. Email: laurence1234kimo{at}yahoo.com.tw. [1]: #xref-corresp-1-1
- Published
- 2019
- Full Text
- View/download PDF
38. STE20/SPS1-related proline/alanine-rich kinase (SPAK) is critical for sodium reabsorption in isolated, perfused thick ascending limb
- Author
-
Joonho Yoon, Michel Baum, Chih-Jen Cheng, and Chou Long Huang
- Subjects
Vasopressins ,Physiology ,Sodium ,chemistry.chemical_element ,In Vitro Techniques ,Protein Serine-Threonine Kinases ,Glutamyl Aminopeptidase ,Loop of Henle ,medicine ,Animals ,Protein Isoforms ,Solute Carrier Family 12, Member 1 ,Mice, Knockout ,Alanine ,Protein-Serine-Threonine Kinases ,Renal sodium reabsorption ,urogenital system ,Kinase ,Chemistry ,Renal Reabsorption ,Articles ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Biochemistry ,Glutamyl aminopeptidase - Abstract
SPAK [STE20 (sterile 20)/SPS1-related proline/alanine-rich kinase] kinase consists of a full-length (FL-) and an alternatively spliced kidney-specific (KS-) isoform. SPAK regulates the NaCl cotransporter (NCC) in the distal convoluted tubule (DCT). The relative abundance and role of FL- vs. KS-SPAK in regulating Na+-K+-2Cl− cotransporter (NKCC2) in thick ascending limb (TAL) are not completely understood. Here, we report that FL-SPAK mRNA was the most abundant in medullary TAL (mTAL), followed by cortical TAL (cTAL) and DCT. KS-SPAK mRNA abundance was relatively lower than FL-SPAK. The ratios of FL-SPAK to KS-SPAK in mTAL, cTAL, and DCT were 12.3, 12.5, and 10.2, respectively. To examine the role of SPAK in the regulation of sodium transport in TAL, we used in vitro microperfusion of mTAL and cTAL isolated from wild-type (WT) and SPAK knockout mice (SPAK-KO) that lack both FL- and KS-SPAK. The rates of sodium absorption in cTAL and mTAL of SPAK-KO mice were 34.5 and 12.5% of WT tubules, respectively. The mRNA levels of related OSR1 kinase and SPAK protease Dnpep in SPAK-KO tubules were not significantly different from WT tubules. We next examined the role of SPAK in the regulation of sodium reabsorption by vasopressin in TAL. Vasopressin increased sodium reabsorption by ∼80% in both mTAL and cTAL from WT mice. While baseline sodium reabsorption was lower in SPAK-KO tubules, vasopressin increased sodium reabsorption over twofold. In conclusion, the combined net effect of SPAK isoforms on sodium reabsorption in TAL is stimulatory. SPAK is not essential for vasopressin stimulation of sodium reabsorption in TAL.
- Published
- 2015
- Full Text
- View/download PDF
39. Potassium-regulated distal tubule WNK bodies are kidney-specific WNK1 dependent
- Author
-
Lubika J. Nkashama, Roderick J. Tan, Donna B. Stolz, Kara L. McClain, Cary R. Boyd-Shiwarski, Thomas R. Kleyman, Hima N. Namboodiri, Manojkumar A. Puthenveedu, Chou Long Huang, Allison L. Marciszyn, Jian Xie, Daniel J. Shiwarski, Ankita Roy, and Arohan R. Subramanya
- Subjects
0301 basic medicine ,Mice, 129 Strain ,030232 urology & nephrology ,Biology ,Kidney ,03 medical and health sciences ,Mice ,0302 clinical medicine ,WNK Lysine-Deficient Protein Kinase 1 ,Organelle ,medicine ,Animals ,Humans ,Distal convoluted tubule ,Microscopy, Immunoelectron ,Molecular Biology ,Mice, Knockout ,urogenital system ,HEK 293 cells ,Kidney metabolism ,Cell Biology ,Exons ,WNK1 ,Cell biology ,Mice, Inbred C57BL ,Cytosol ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Potassium ,Signal transduction ,Hydrophobic and Hydrophilic Interactions ,Homeostasis ,Signal Transduction - Abstract
With-no-lysine (WNK) kinases coordinate volume and potassium homeostasis by regulating renal tubular electrolyte transport. In the distal convoluted tubule (DCT), potassium imbalance causes WNK signaling complexes to concentrate into large discrete foci, which we call “WNK bodies.” Although these structures have been reported previously, the mechanisms that drive their assembly remain obscure. Here, we show that kidney-specific WNK1 (KS-WNK1), a truncated kinase-defective WNK1 isoform that is highly expressed in the DCT, is critical for WNK body formation. While morphologically distinct WNK bodies were evident in the distal tubules of mice subjected to dietary potassium loading and restriction, KS-WNK1 knockout mice were deficient in these structures under identical conditions. Combining in vivo observations in kidney with reconstitution studies in cell culture, we found that WNK bodies are dynamic membraneless foci that are distinct from conventional organelles, colocalize with the ribosomal protein L22, and cluster the WNK signaling pathway. The formation of WNK bodies requires an evolutionarily conserved cysteine-rich hydrophobic motif harbored within a unique N-terminal exon of KS-WNK1. We propose that WNK bodies are not pathological aggregates, but rather are KS-WNK1–dependent microdomains of the DCT cytosol that modulate WNK signaling during physiological shifts in potassium balance.
- Published
- 2017
40. Modeled structural basis for the recognition of α2-3-sialyllactose by soluble Klotho
- Author
-
Jon D. Wright, Noelynn Oliver, Nicole Nischan, Joonho Yoon, Sung Wan An, Jennifer J. Kohler, Jian Xie, Chou Long Huang, and Carmay Lim
- Subjects
0301 basic medicine ,Models, Molecular ,Protein Conformation ,Cardiomegaly ,Lactose ,Biochemistry ,Exocytosis ,TRPC6 ,03 medical and health sciences ,Transient receptor potential channel ,Mice ,Phosphatidylinositol 3-Kinases ,Membrane Microdomains ,Cell surface receptor ,Gangliosides ,Genetics ,Carbohydrate Conformation ,Animals ,Humans ,Computer Simulation ,Homology modeling ,Amino Acid Sequence ,Molecular Biology ,Lipid raft ,Klotho ,Klotho Proteins ,Glucuronidase ,Binding Sites ,030102 biochemistry & molecular biology ,Chemistry ,Research ,Cell biology ,030104 developmental biology ,HEK293 Cells ,Ectodomain ,Gene Expression Regulation ,Sialic Acids ,Biotechnology ,Signal Transduction - Abstract
Soluble Klotho (sKlotho) is the shed ectodomain of antiaging membrane Klotho that contains 2 extracellular domains KL1 and KL2, each of which shares sequence homology to glycosyl hydrolases. sKlotho elicits pleiotropic cellular responses with a poorly understood mechanism of action. Notably, in injury settings, sKlotho confers cardiac and renal protection by down-regulating calcium-permeable transient receptor potential canonical type isoform 6 (TRPC6) channels in cardiomyocytes and glomerular podocytes. Inhibition of PI3K-dependent exocytosis of TRPC6 is thought to be the underlying mechanism, and recent studies showed that sKlotho interacts with α2-3-sialyllactose-containing gangliosides enriched in lipid rafts to inhibit raft-dependent PI3K signaling. However, the structural basis for binding and recognition of α2-3-sialyllactose by sKlotho is unknown. Using homology modeling followed by docking, we identified key protein residues in the KL1 domain that are likely involved in binding sialyllactose. Functional experiments based on the ability of Klotho to down-regulate TRPC6 channel activity confirm the importance of these residues. Furthermore, KL1 domain binds α2-3-sialyllactose, down-regulates TRPC6 channels, and exerts protection against stress-induced cardiac hypertrophy in mice. Our results support the notion that sialogangliosides and lipid rafts are membrane receptors for sKlotho and that the KL1 domain is sufficient for the tested biologic activities. These findings can help guide the design of a simpler Klotho mimetic.-Wright, J. D., An, S.-W., Xie, J., Yoon, J., Nischan, N., Kohler, J. J., Oliver, N., Lim, C., Huang, C.-L. Modeled structural basis for the recognition of α2-3-sialyllactose by soluble Klotho.
- Published
- 2017
41. Soluble klotho binds monosialoganglioside to regulate membrane microdomains and growth factor signaling
- Author
-
George D. Dalton, Nicole Nischan, Evgenia Dobrinskikh, Joonho Yoon, Donald W. Hilgemann, Jennifer J. Kohler, Lutz Birnbaumer, Katherine Luby-Phelps, Saif I. Al-Juboori, Jian Xie, Sung Wan An, and Chou Long Huang
- Subjects
0301 basic medicine ,GANGLIOSIDES ,CIENCIAS MÉDICAS Y DE LA SALUD ,Inmunología ,TRPC6 ,Biology ,urologic and male genital diseases ,Endocytosis ,Biophysical Phenomena ,LIPIDOS ,Cell Line ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,Paracrine signalling ,Membrane Microdomains ,Cell Line, Tumor ,Gangliosides ,Animals ,Humans ,Klotho Proteins ,Protein kinase B ,Lipid raft ,Klotho ,PI3K/AKT/mTOR pathway ,SOLUBLE KLOTHO ,Glucuronidase ,TUMORES ,MEMBRANAS CELULARES ,Multidisciplinary ,030102 biochemistry & molecular biology ,SIALIC ACIDS ,PROTEINAS ,purl.org/becyt/ford/3.1 [https] ,Biological Sciences ,humanities ,female genital diseases and pregnancy complications ,Cell biology ,Medicina Básica ,LIPID RAFTS ,HEK293 Cells ,030104 developmental biology ,Ectodomain ,Membrane protein ,Intercellular Signaling Peptides and Proteins ,lipids (amino acids, peptides, and proteins) ,purl.org/becyt/ford/3 [https] ,HeLa Cells ,Signal Transduction - Abstract
Fil: Dalton, George. University of Texas Southwestern Medical Center. Department of Medicine; Estados Unidos Fil: An, Sung-Wan. University of Texas Southwestern Medical Center. Department of Medicine; Estados Unidos Fil: Al-Juboori, Saif I. University of Colorado Denver. Department of Electrical Engineering; Estados Unidos Fil: Nischan, Nicole. University of Texas Southwestern Medical Center. Department of Biochemistry; Estados Unidos Fil: Yoon, Joonho. University of Texas Southwestern Medical Center. Department of Medicine; Estados Unidos Fil: Dobrinskikh, Evgenia. University of Colorado Denver. Department of Medicine; Estados Unidos Fil: Hilgemann, Donald W. University of Texas Southwestern Medical Center. Department of Physiology; Estados Unidos Fil: Xie, Jian. University of Texas Southwestern Medical Center. Department of Medicine; Estados Unidos Fil: Luby-Phelps, Kate. University of Texas Southwestern Medical Center. Department of Cell Biology; Estados Unidos Fil: Luby-Phelps, Kate. University of Texas Southwestern Medical Center. Live Cell Imaging Core Facility; Estados Unidos Fil: Kohler, Jennifer J. University of Texas Southwestern Medical Center. Department of Biochemistry; Estados Unidos Fil: Birnbaumer, Lutz. Pontificia Universidad Católica Argentina. Facultad de Ciencias Médicas. Instituto de Investigaciones Biomédicas; Argentina Fil: Birnbaumer, Lutz. National Institute of Environmental Health Sciences. Neurobiology Laboratory; Estados Unidos Fil: Huang, Chou-Long. University of Texas Southwestern Medical Center. Department of Medicine; Estados Unidos Abstract: Soluble klotho, the shed ectodomain of the antiaging membrane protein α-klotho, is a pleiotropic endocrine/paracrine factor with no known receptors and poorly understood mechanism of action. Soluble klotho down-regulates growth factor-driven PI3K signaling, contributing to extension of lifespan, cardioprotection, and tumor inhibition. Here we show that soluble klotho binds membrane lipid rafts. Klotho binding to rafts alters lipid organization, decreases membrane's propensity to form large ordered domains for endocytosis, and down-regulates raft-dependent PI3K/Akt signaling. We identify α2-3-sialyllactose present in the glycan of monosialogangliosides as targets of soluble klotho. α2-3-Sialyllactose is a common motif of glycans. To explain why klotho preferentially targets lipid rafts we show that clustering of gangliosides in lipid rafts is important. In vivo, raft-dependent PI3K signaling is up-regulated in klotho-deficient mouse hearts vs. wild-type hearts. Our results identify ganglioside-enriched lipid rafts to be receptors that mediate soluble klotho regulation of PI3K signaling. Targeting sialic acids may be a general mechanism for pleiotropic actions of soluble klotho.
- Published
- 2017
42. An acetate switch regulates stress erythropoiesis
- Author
-
Holly Walters, Rui Chen, Young Ah Moon, Min Xu, Jason S. Nagati, Jiwen Li, Robert D. Gerard, Sarah A. Comerford, Joseph A. Garcia, Jian Xie, Jay D. Horton, Robert E. Hammer, and Chou Long Huang
- Subjects
medicine.medical_specialty ,Anemia ,p300 ,Acetates ,Biology ,Cbp ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Stress, Physiological ,hemic and lymphatic diseases ,Internal medicine ,ACSS2 ,Acss2 ,medicine ,Animals ,Erythropoiesis ,Erythropoietin ,Transcription factor ,Regulation of gene expression ,HIF-2 ,fungi ,food and beverages ,Acetylation ,General Medicine ,medicine.disease ,Cell Hypoxia ,Endocrinology ,Signal transduction ,Signal Transduction ,Transcription Factors ,Epo ,medicine.drug - Abstract
The hormone erythropoietin (EPO), which is synthesized in the kidney or liver of adult mammals, controls erythrocyte production and is regulated by the stress-responsive transcription factor hypoxia-inducible factor-2 (HIF-2). We previously reported that the lysine acetyltransferase CREB-binding protein (CBP) is required for HIF-2α acetylation and efficient HIF-2-dependent EPO induction during hypoxia. We now show that these processes require acetate-dependent acetyl CoA synthetase 2 (ACSS2). In human Hep3B hepatoma cells and in EPO-generating organs of hypoxic or acutely anemic mice, acetate levels rise and ACSS2 is required for HIF-2α acetylation, CBP-HIF-2α complex formation, CBP-HIF-2α recruitment to the EPO enhancer and efficient induction of EPO gene expression. In acutely anemic mice, acetate supplementation augments stress erythropoiesis in an ACSS2-dependent manner. Moreover, in acquired and inherited chronic anemia mouse models, acetate supplementation increases EPO expression and the resting hematocrit. Thus, a mammalian stress-responsive acetate switch controls HIF-2 signaling and EPO induction during pathophysiological states marked by tissue hypoxia.
- Published
- 2014
- Full Text
- View/download PDF
43. WNK1-OSR1/SPAK KINASE CASCADE IS IMPORTANT FOR ANGIOGENESIS.
- Author
-
CHOU-LONG HUANG, JIAN XIE, and CHIOU-HWA YUH
- Subjects
SERINE/THREONINE kinases ,GENETIC mutation ,OXIDATIVE stress ,ENDOTHELIUM ,TRANSGENES ,ENDOTHELIAL cells - Abstract
WNK [with-no-lysine (K)] kinases are a family of four members of serine and threonine kinases that regulate renal Na+ and K+ transport. Mutations of WNK1 and WNK4 cause a hereditary hypertensive and hyperkalemic disease known as pseudohypoaldosteronism type II (PHA2). Unlike other WNK isoforms, WNK1 is ubiquitously expressed and regulates many other cellular processes outside the kidney. Oxidative stress response kinase (OSR1) and related STE 20/SPS1-related proline alanine-rich kinase (SPAK) are downstream kinases of WNK kinases. To examine the role of WNK kinase cascade in vivo, we generated global Wnk1-deleted mice and found that Wnk1-ablated mice die in utero from embryonic angiogenesis and cardiac developmental defects. Endothelial-specific Wnk1 deletion reveals that angiogenesis defect is due to WNK1 requirement in endothelium. We further showed that global and endothelial-deletion of Osr1 phenocopies Wnk1 deletion. Furthermore, expression of a catalytic constitutively active Osr1 transgene rescues angiogenesis defects and embryonic lethality of Wnk1-ablated mice. In zebrafish, Wnk1 knockdown causes similar angiogenesis defects to Vegf2 (Flk1) knockdown and that expression of WNK1 partially rescues Flk1 angiogenesis defects. The results indicate that WNK1 is downstream of VEGF signaling cascade. T-lymphocytes isolated from Wnk1-null mice exhibit migration defects. Inhibition of WNK1-OSR1 downstream target Na-K-2Cl cotransporter NKCC1 mimics migration defect of WNK1-deficient T-lymphocytes. Thus, WNK1-OSR1/SPAK cascade is important for angiogenesis. Regulation of ion homeostasis and cell volume may underlie the mechanism for WNK1 regulation of endothelial cell migration and angiogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
44. Acquired Hypocalciuric Hypercalcemia in a Patient With CKD
- Author
-
Anthony P. Weetman, Harpreet K. Sandhu, E. Helen Kemp, Elizabeth Kuo, Edward M. Brown, and Chou Long Huang
- Subjects
medicine.medical_specialty ,Cinacalcet ,endocrine system diseases ,Calcimimetic ,Hypocalciuria ,Autoimmune Diseases ,Internal medicine ,medicine ,Humans ,Autoantibodies ,Aged, 80 and over ,Hyperparathyroidism ,business.industry ,medicine.disease ,Urinary calcium ,Endocrinology ,Nephrology ,Hypercalcemia ,Kidney Failure, Chronic ,Calcium ,Female ,Hyperparathyroidism, Secondary ,medicine.symptom ,Calcium-sensing receptor ,business ,Receptors, Calcium-Sensing ,hormones, hormone substitutes, and hormone antagonists ,Primary hyperparathyroidism ,medicine.drug ,Kidney disease - Abstract
We present a case of an 82-year-old woman with elevated parathyroid hormone (PTH) levels, hypocalciuria, hypercalcemia, and stage 3 chronic kidney disease. Hypocalciuria initially was attributed to chronic kidney disease, and hypercalcemia was attributed to primary hyperparathyroidism. Subsequent laboratory studies showed autoantibodies in the patient's serum directed against the calcium-sensing receptor (CaSR). Functional testing in a CaSR-transfected human embryonic kidney-293 cell line showed that the patient's antibodies inhibited CaSR-mediated intracellular signaling that ordinarily would have been stimulated by extracellular calcium ions. Her serum calcium and PTH levels were normalized by treatment with the calcimimetic cinacalcet. We advise consideration of the presence of inhibitory autoantibodies directed at the CaSR in patients with hypercalcemic hyperparathyroidism and unexplained hypocalciuria or with confounding conditions affecting interpretation of urinary calcium measurement. A calcimimetic is an effective treatment for the hypercalcemia and elevated PTH levels in acquired hypocalciuric hypercalcemia caused by inhibitory anti-CaSR autoantibodies.
- Published
- 2013
- Full Text
- View/download PDF
45. Flow-induced activation of TRPV5 and TRPV6 channels stimulates Ca2+-activated K+ channel causing membrane hyperpolarization
- Author
-
Chou Long Huang, Seung Kuy Cha, and Ji Hee Kim
- Subjects
BK channel ,Glycosylation ,TRPV Cation Channels ,Flow-mediated Ca2+ entry ,Article ,Membrane Potentials ,SK channel ,Potassium Channels, Calcium-Activated ,Flow-mediated K+ secretion ,Humans ,Large-Conductance Calcium-Activated Potassium Channels ,Potassium Channels, Inwardly Rectifying ,ROMK ,Molecular Biology ,Membrane potential ,biology ,Voltage-gated ion channel ,Reabsorption ,Chemistry ,Cardiac action potential ,Membrane hyperpolarization ,Cell Biology ,Hyperpolarization (biology) ,HEK293 Cells ,Biochemistry ,biology.protein ,Biophysics ,Calcium ,Stress, Mechanical ,Rheology ,TRPV5 ,Ion Channel Gating ,TRPV6 ,Ca2+-activated K+ channel - Abstract
TRPV5 and TRPV6 channels are expressed in distal renal tubules and play important roles in the transcellular Ca 2 + reabsorption in kidney. They are regulated by multiple intracellular factors including protein kinases A and C, membrane phospholipid PIP 2 , protons, and divalent ions Ca 2 + and Mg 2 + . Here, we report that fluid flow that generates shear force within the physiological range of distal tubular fluid flow activated TRPV5 and TRPV6 channels expressed in HEK cells. Flow-induced activation of channel activity was reversible and did not desensitize over 2 min. Fluid flow stimulated TRPV5 and 6-mediated Ca 2 + entry and increased intracellular Ca 2 + concentration. N-glycosylation-deficient TRPV5 channel was relatively insensitive to fluid flow. In cells coexpressing TRPV5 (or TRPV6) and Slo1 -encoded maxi-K channels, fluid flow induced membrane hyperpolarization, which could be prevented by the maxi-K blocker iberiotoxin or TRPV5 and 6 blocker La 3 + . In contrast, fluid flow did not cause membrane hyperpolarization in cells coexpressing ROMK1 and TRPV5 or 6 channel. These results reveal a new mechanism for the regulation of TRPV5 and TRPV6 channels. Activation of TRPV5 and TRPV6 by fluid flow may play a role in the regulation of flow-stimulated K + secretion via maxi-K channels in distal renal tubules and in the mechanism of pathogenesis of thiazide-induced hypocalciuria.
- Published
- 2013
- Full Text
- View/download PDF
46. DGKE Variants Cause a Glomerular Microangiopathy That Mimics Membranoproliferative GN
- Author
-
Nesrin Besbas, Nicolas G. Bazan, Richard F. Ransom, Chaoying Liang, Edward K. Wakeland, Aysin Bakkaloglu, Tulin Ibsirlioglu, Fatih Ozaltin, Massimo Attanasio, Benjamin E. Wakeland, Komal Vadnagara, Ali Duzova, Emine Korkmaz, Sung Wan An, Richard J. Quigg, Chou Long Huang, Yong Du, Shazia Ashraf, Oguz Soylemezoglu, Alysha Rauhauser, Chandra Mohan, Binghua Li, Deborah Lewis, Friedhelm Hildebrandt, Dongmei Lu, Ekim Z. Taskiran, Rezan Topaloglu, Yelda Bilginer, Phylip Chen, Susan Arbuckle, Seza Ozen, İpek Işık Gönül, Matthew K. Topham, and Çocuk Sağlığı ve Hastalıkları
- Subjects
Male ,Diacylglycerol Kinase ,Turkey ,Glomerulonephritis, Membranoproliferative ,Kidney Glomerulus ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,Podocyte ,Cohort Studies ,Diagnosis, Differential ,Diglycerides ,Consanguinity ,Mice ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Exome sequencing ,Diacylglycerol kinase ,Genetics ,Mutation ,Base Sequence ,Sequence Homology, Amino Acid ,Podocytes ,Kinase ,Microangiopathy ,HEK 293 cells ,Genetic Variation ,Glomerulonephritis ,DNA ,General Medicine ,Urology & Nephrology ,medicine.disease ,Molecular biology ,Pedigree ,Rats ,HEK293 Cells ,medicine.anatomical_structure ,Nephrology ,Female ,Kidney Diseases - Abstract
Renal microangiopathies and membranoproliferative GN (MPGN) can manifest similar clinical presentations and histology, suggesting the possibility of a common underlying mechanism in some cases. Here, we performed homozygosity mapping and whole exome sequencing in a Turkish consanguineous family and identified DGKE gene variants as the cause of a membranoproliferative-like glomerular microangiopathy. Furthermore, we identified two additional DGKE variants in a cohort of 142 unrelated patients diagnosed with membranoproliferative GN. This gene encodes the diacylglycerol kinase DGK epsilon, which is an intracellular lipid kinase that phosphorylates diacylglycerol to phosphatidic acid. Immunofluorescence confocal microscopy demonstrated that mouse and rat Dgk epsilon colocalizes with the podocyte marker WT1 but not with the endothelial marker CD31. Patch-clamp experiments in human embryonic kidney (HEK293) cells showed that DGK epsilon variants affect the intracellular concentration of diacylglycerol. Taken together, these results not only identify a genetic cause of a glomerular microangiopathy but also suggest that the phosphatidylinositol cycle, which requires DGKE, is critical to the normal function of podocytes. J Am Soc Nephrol 24: 377-384, 2013. doi: 10.1681/ASN.2012090903
- Published
- 2013
- Full Text
- View/download PDF
47. Kidney-specific WNK1 regulates sodium reabsorption and potassium secretion in mouse cortical collecting duct
- Author
-
Chou Long Huang, Michel Baum, and Chih-Jen Cheng
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Sodium ,chemistry.chemical_element ,Protein Serine-Threonine Kinases ,Biology ,Absorption ,Membrane Potentials ,Minor Histocompatibility Antigens ,Tissue Culture Techniques ,Mice ,WNK Lysine-Deficient Protein Kinase 1 ,Internal medicine ,medicine ,Animals ,Large-Conductance Calcium-Activated Potassium Channels ,Distal convoluted tubule ,Kidney Tubules, Collecting ,Potassium Channels, Inwardly Rectifying ,Kidney Tubules, Distal ,Sequence Deletion ,Transepithelial potential difference ,Mice, Knockout ,Renal sodium reabsorption ,urogenital system ,Reabsorption ,Potassium, Dietary ,Exons ,Articles ,Iberiotoxin ,equipment and supplies ,Potassium channel ,Connecting tubule ,Perfusion ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Potassium ,Female ,Peptides - Abstract
Kidney-specific with-no-lysine kinase 1 (KS-WNK1) is a kinase-deficient variant of WNK1 that is expressed exclusively in the kidney. It is abundantly expressed in the distal convoluted tubule (DCT) and to a lesser extent in the cortical thick ascending limb (cTAL), connecting tubule, and cortical collecting duct (CCD). KS-WNK1 inhibits Na+-K+-2Cl−- and sodium chloride cotransporter-mediated Na+reabsorption in cTAL and DCT, respectively. Here, we investigated the role of KS-WNK1 in regulating Na+and K+transport in CCD using in vitro microperfusion of tubules isolated from KS-WNK1 knockout mice and control wild-type littermates. Because baseline K+secretion and Na+reabsorption were negligible in mouse CCD, we studied tubules isolated from mice fed a high-K+diet for 2 wk. Compared with that in wild-type tubules, K+secretion was reduced in KS-WNK1 knockout CCD perfused at a low luminal fluid rate of ∼1.5 nl/min. Na+reabsorption and the lumen-negative transepithelial potential difference were also lower in the KS-WNK1 knockout CCD compared with control CCD. Increasing the perfusion rate to ∼5.5 nl/min stimulated K+secretion in the wild-type as well as knockout CCD. The magnitudes of flow-stimulated increase in K+secretion were similar in wild-type and knockout CCD. Maxi-K+channel inhibitor iberiotoxin had no effect on K+secretion when tubules were perfused at ∼1.5 nl/min, but completely abrogated the flow-dependent increase in K+secretion at ∼5.5 nl/min. These findings support the notion that KS-WNK1 stimulates ROMK-mediated K+secretion, but not flow-dependent K+secretion mediated by maxi-K+channels in CCD. In addition, KS-WNK1 plays a role in regulating Na+transport in the CCD.
- Published
- 2013
- Full Text
- View/download PDF
48. High dietary phosphate intake induces hypertension and augments exercise pressor reflex function in rats
- Author
-
Ming Chang Hu, Naim M. Maalouf, Scott A. Smith, Orson W. Moe, Masaki Mizuno, Chou Long Huang, Scott Crawford, Jere H. Mitchell, and Wanpen Vongpatanasin
- Subjects
medicine.medical_specialty ,Contraction (grammar) ,Sympathetic Nervous System ,Physiology ,Physical Exertion ,030232 urology & nephrology ,Blood Pressure ,Hindlimb ,030204 cardiovascular system & hematology ,Kidney ,Phosphates ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Heart Rate ,Physiology (medical) ,Internal medicine ,medicine ,Pi ,Animals ,Exertion ,Muscle, Skeletal ,Neural Control ,business.industry ,Skeletal muscle ,Baroreflex ,Diet ,Rats ,medicine.anatomical_structure ,Blood pressure ,Endocrinology ,chemistry ,Capsaicin ,Hypertension ,Reflex ,business ,Muscle Contraction - Abstract
An increasing number of studies have linked high dietary phosphate (Pi) intake to hypertension. It is well established that the rise in sympathetic nerve activity (SNA) and blood pressure (BP) during physical exertion is exaggerated in many forms of hypertension, which are primarily mediated by an overactive skeletal muscle exercise pressor reflex (EPR). However, it remains unknown whether high dietary Pi intake potentiates the EPR-mediated SNA and BP response to exercise. Accordingly, we measured renal SNA (RSNA) and mean BP (MBP) in normotensive Sprague-Dawley rats fed a normal Pi diet (0.6%, n = 13) or high Pi diet (1.2%, n = 13) for 3 mo. As previously reported, we found that resting BP was significantly increased by 1.2% Pi diet in both conscious and anesthetized animals. Activation of the EPR by electrically induced hindlimb contraction triggered greater increases in ΔRSNA and ΔMBP in the 1.2% compared with 0.6% Pi group (126 ± 25 vs. 42 ± 9%; 44 ± 5 vs. 14 ± 2 mmHg, respectively, P < 0.01). Activation of the muscle mechanoreflex, a component of the EPR, by passively stretching hindlimb muscle also evoked greater increases in ΔRSNA and ΔMBP in the 1.2% compared with 0.6% Pi group (109 ± 27 vs. 24 ± 7%, 38 ± 7 vs. 8 ± 2 mmHg, respectively, P < 0.01). A similar response was produced by hindlimb intra-arterial capsaicin administration to stimulate the metaboreflex arm of the EPR. Thus, our data demonstrate a novel action of dietary Pi loading in augmenting EPR function through overactivation of both the muscle mechanoreflex and metaboreflex.
- Published
- 2016
49. Klotho May Ameliorate Proteinuria by Targeting TRPC6 Channels in Podocytes
- Author
-
Chou Long Huang, Minseob Eom, Seung Kuy Cha, Kyu Hee Hwang, Yueh Lin Wu, Kyu Sang Park, Noelynn Oliver, Nestor X. Barrezueta, Ji Hee Kim, Jian Xie, R. Paul Fracasso, and In Deok Kong
- Subjects
0301 basic medicine ,medicine.medical_specialty ,In situ hybridization ,Biology ,urologic and male genital diseases ,TRPC6 ,Podocyte ,03 medical and health sciences ,Transient receptor potential channel ,Mice ,Internal medicine ,Extracellular ,medicine ,TRPC6 Cation Channel ,Albuminuria ,Animals ,Humans ,Renal Insufficiency, Chronic ,Klotho ,Klotho Proteins ,Cells, Cultured ,Glucuronidase ,TRPC Cation Channels ,Kidney ,Podocytes ,General Medicine ,female genital diseases and pregnancy complications ,Proteinuria ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Basic Research ,Nephrology ,medicine.symptom - Abstract
Klotho is a type-1 membrane protein predominantly produced in the kidney, the extracellular domain of which is secreted into the systemic circulation. Membranous and secreted Klotho protect organs, including the kidney, but whether and how Klotho directly protects the glomerular filter is unknown. Here, we report that secreted Klotho suppressed transient receptor potential channel 6 (TRPC6)-mediated Ca2+ influx in cultured mouse podocytes by inhibiting phosphoinositide 3-kinase-dependent exocytosis of the channel. Furthermore, soluble Klotho reduced ATP-stimulated actin cytoskeletal remodeling and transepithelial albumin leakage in these cells. Overexpression of TRPC6 by gene delivery in mice induced albuminuria, and exogenous administration of Klotho ameliorated the albuminuria. Notably, immunofluorescence and in situ hybridization revealed Klotho expression in podocytes of mouse and human kidney. Heterozygous Klotho-deficient CKD mice had aggravated albuminuria compared with that in wild-type CKD mice with a similar degree of hypertension and reduced clearance function. Finally, disrupting the integrity of glomerular filter by saline infusion-mediated extracellular fluid volume expansion increased urinary Klotho excretion. These results reveal a potential novel function of Klotho in protecting the glomerular filter, and may offer a new therapeutic strategy for treatment of proteinuria.
- Published
- 2016
50. Hypertension: the missing WNKs
- Author
-
Melanie H. Cobb, Hashem A. Dbouk, and Chou Long Huang
- Subjects
0301 basic medicine ,Physiology ,Reviews ,Blood Pressure ,Protein Serine-Threonine Kinases ,Pharmacology ,Sodium Chloride ,03 medical and health sciences ,medicine ,Animals ,Humans ,Sodium Chloride, Dietary ,chemistry.chemical_classification ,Kidney ,Protein-Serine-Threonine Kinases ,business.industry ,urogenital system ,Intracellular Signaling Peptides and Proteins ,Cell biology ,030104 developmental biology ,Blood pressure ,Enzyme ,medicine.anatomical_structure ,chemistry ,Hypertension ,Signal transduction ,business ,Cotransporter ,Signal Transduction - Abstract
The With no Lysine [K] (WNK) family of enzymes are central in the regulation of blood pressure. WNKs have been implicated in hereditary hypertension disorders, mainly through control of the activity and levels of ion cotransporters and channels. Actions of WNKs in the kidney have been heavily investigated, and recent studies have provided insight into not only the regulation of these enzymes but also how mutations in WNKs and their interacting partners contribute to hypertensive disorders. Defining the roles of WNKs in the cardiovascular system will provide clues about additional mechanisms by which WNKs can regulate blood pressure. This review summarizes recent developments in the regulation of the WNK signaling cascade and its role in regulation of blood pressure.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.