1
|
Rodan AR. With No Lysine (K) Kinases and Sodium Transporter Function in Solute Exchange with Implications for BP Regulation as Elucidated through Drosophila. KIDNEY360 2024; 5:1553-1562. [PMID: 39186374 DOI: 10.34067/kid.0000000000000564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Like other multicellular organisms, the fruit fly Drosophila melanogaster must maintain homeostasis of the internal milieu, including the maintenance of constant ion and water concentrations. In mammals, the with no lysine (K) (WNK)-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade is an important regulator of epithelial ion transport in the kidney. This pathway regulates SLC12 family cotransporters, including sodium-potassium-2-chloride, sodium chloride, and potassium chloride cotransporters. The WNK-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade also regulates epithelial ion transport via regulation of the Drosophila sodium-potassium-2-chloride cotransporter in the Malpighian tubule, the renal epithelium of the fly. Studies in Drosophila have contributed to the understanding of multiple regulators of WNK pathway signaling, including intracellular chloride and potassium, the scaffold protein Mo25, hypertonic stress, hydrostatic pressure, and macromolecular crowding. These will be discussed together, with implications for mammalian kidney function and BP control.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah; Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah; Department of Human Genetics, University of Utah, Salt Lake City, Utah; and Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| |
Collapse
|
2
|
Kim JS, Kehrl JH. Inhibition of WNK Kinases in NK Cells Disrupts Cellular Osmoregulation and Control of Tumor Metastasis. J Innate Immun 2024; 16:451-469. [PMID: 39265537 PMCID: PMC11521464 DOI: 10.1159/000540744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 08/01/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION The serine/threonine with-no-lysine (WNK) kinase family function in blood pressure control, electrolyte homeostasis, and cellular osmoregulation. These kinases and their downstream effectors are considered promising therapeutic targets in hypertension and stroke. However, the role of WNK kinases in immune cells remains poorly understood. METHODS Using the small-molecule WNK kinase inhibitors WNK463 and WNK-IN-11, we investigated how WNK kinase inhibition affects natural killer (NK) cell physiology. RESULTS WNK kinase inhibition with WNK463 or WNK-IN-11 significantly decreased IL-2-activated NK cell volume, motility, and cytolytic activity. Treatment of NK cells with these inhibitors induced autophagy by activating AMPK and inhibiting mTOR signaling. Moreover, WNK kinase inhibition increased phosphorylation of Akt and c-Myc by misaligning activity of activating kinases and inhibitory phosphatases. Treatment of tumor-bearing mice with WNK463 impaired tumor metastasis control by adoptively transferred NK cells. CONCLUSION The catalytic activity of WNK kinases has a critical role of multiple aspects of NK cell physiology and their pharmacologic inhibition negatively impacts NK cell function.
Collapse
Affiliation(s)
- Ji Sung Kim
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Boyd-Shiwarski CR, Shiwarski DJ, Subramanya AR. A New Phase for WNK Kinase Signaling Complexes as Biomolecular Condensates. Physiology (Bethesda) 2024; 39:0. [PMID: 38624245 PMCID: PMC11460533 DOI: 10.1152/physiol.00013.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
The purpose of this review is to highlight transformative advances that have been made in the field of biomolecular condensates, with special emphasis on condensate material properties, physiology, and kinases, using the With-No-Lysine (WNK) kinases as a prototypical example. To convey how WNK kinases illustrate important concepts for biomolecular condensates, we start with a brief history, focus on defining features of biomolecular condensates, and delve into some examples of how condensates are implicated in cellular physiology (and pathophysiology). We then highlight how WNK kinases, through the action of "WNK droplets" that ubiquitously regulate intracellular volume and kidney-specific "WNK bodies" that are implicated in distal tubule salt reabsorption and potassium homeostasis, exemplify many of the defining features of condensates. Finally, this review addresses the controversies within this emerging field and questions to address.
Collapse
Affiliation(s)
- Cary R Boyd-Shiwarski
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Daniel J Shiwarski
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
4
|
Omage K, McCormick JA. Cullin 3/with No Lysine [K] Kinase/Ste20/SPS-Related Proline Alanine Rich Kinase Signaling: Impact on NaCl Cotransporter Activity in BP Regulation. KIDNEY360 2024; 5:1386-1393. [PMID: 39120943 PMCID: PMC11441819 DOI: 10.34067/kid.0000000000000527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 08/11/2024]
Abstract
The sodium chloride cotransporter (NCC) fine-tunes Na + balance and indirectly affects the homeostasis of other ions including K + , Mg 2+ , and Ca 2+ . Owing to its effects on Na + balance, BP is significantly affected by alterations in NCC activity. Several factors have been reported to influence the expression and activity of NCC. One critical factor is NCC phosphorylation/dephosphorylation that occurs at key serine-threonine amino acid residues of the protein. Phosphorylation, which results in increased NCC activity, is mediated by the with no lysine [K] (WNK)-SPS-related proline alanine rich kinase (SPAK)/OSR1 kinases. NCC activation stimulates reabsorption of Na + , increasing extracellular fluid volume and hence BP. On the other hand, proteasomal degradation of WNK kinases after ubiquitination by the Cullin 3-Kelch-like 3 E3 ubiquitin ligase complex and dephosphorylation pathways oppose WNK-SPAK/OSR1-mediated NCC activation. Components of the Cullin 3/Kelch-like 3-WNK-SPAK/OSR1 regulatory pathway may be targets for novel antihypertensive drugs. In this review, we outline the impact of these regulators on the activity of NCC and the consequent effect on BP.
Collapse
Affiliation(s)
- Kingsley Omage
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
5
|
Ye T, Mishra AK, Banday S, Li R, Hu K, Coleman MM, Shan Y, Chowdhury SR, Zhou L, Pak ML, Simone TM, Malonia SK, Zhu LJ, Kelliher MA, Green MR. Identification of WNK1 as a therapeutic target to suppress IgH/MYC expression in multiple myeloma. Cell Rep 2024; 43:114211. [PMID: 38722741 DOI: 10.1016/j.celrep.2024.114211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/10/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Multiple myeloma (MM) remains an incurable hematological malignancy demanding innovative therapeutic strategies. Targeting MYC, the notorious yet traditionally undruggable oncogene, presents an appealing avenue. Here, using a genome-scale CRISPR-Cas9 screen, we identify the WNK lysine-deficient protein kinase 1 (WNK1) as a regulator of MYC expression in MM cells. Genetic and pharmacological inhibition of WNK1 reduces MYC expression and, further, disrupts the MYC-dependent transcriptional program. Mechanistically, WNK1 inhibition attenuates the activity of the immunoglobulin heavy chain (IgH) enhancer, thus reducing MYC transcription when this locus is translocated near the MYC locus. WNK1 inhibition profoundly impacts MM cell behaviors, leading to growth inhibition, cell-cycle arrest, senescence, and apoptosis. Importantly, the WNK inhibitor WNK463 inhibits MM growth in primary patient samples as well as xenograft mouse models and exhibits synergistic effects with various anti-MM compounds. Collectively, our study uncovers WNK1 as a potential therapeutic target in MM.
Collapse
Affiliation(s)
- Tianyi Ye
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Alok K Mishra
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shahid Banday
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Rui Li
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kai Hu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Madison M Coleman
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Yi Shan
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shreya Roy Chowdhury
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lin Zhou
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Magnolia L Pak
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tessa M Simone
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sunil K Malonia
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Michelle A Kelliher
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Michael R Green
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
6
|
Sharma P, Chatrathi HE. Insights into the diverse mechanisms and effects of variant CUL3-induced familial hyperkalemic hypertension. Cell Commun Signal 2023; 21:286. [PMID: 37845702 PMCID: PMC10577937 DOI: 10.1186/s12964-023-01269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/12/2023] [Indexed: 10/18/2023] Open
Abstract
Familial hyperkalemic hypertension (FHHt), also known as Pseudohypoaldosteronism type II (PHAII) or Gordon syndrome is a rare Mendelian disease classically characterized by hyperkalemia, hyperchloremic metabolic acidosis, and high systolic blood pressure. The most severe form of the disease is caused by autosomal dominant variants in CUL3 (Cullin 3), a critical subunit of the multimeric CUL3-RING ubiquitin ligase complex. The recent identification of a novel FHHt disease variant of CUL3 revealed intricacies within the underlying disease mechanism. When combined with studies on canonical CUL3 variant-induced FHHt, these findings further support CUL3's role in regulating renal electrolyte transport and maintaining systemic vascular tone. However, the pathophysiological effects of CUL3 variants are often accompanied by diverse systemic disturbances in addition to classical FHHt symptoms. Recent global proteomic analyses provide a rationale for these systemic disturbances, paving the way for future mechanistic studies to reveal how CUL3 variants dysregulate processes outside of the renovascular axis. Video Abstract.
Collapse
Affiliation(s)
- Prashant Sharma
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA.
| | - Harish E Chatrathi
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
7
|
Humphreys JM, Teixeira LR, Akella R, He H, Kannangara AR, Sekulski K, Pleinis J, Liwocha J, Jiou J, Servage KA, Orth K, Joachimiak L, Rizo J, Cobb MH, Brautigam CA, Rodan AR, Goldsmith EJ. Hydrostatic Pressure Sensing by WNK kinases. Mol Biol Cell 2023; 34:ar109. [PMID: 37585288 PMCID: PMC10559305 DOI: 10.1091/mbc.e23-03-0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
Previous study has demonstrated that the WNK kinases 1 and 3 are direct osmosensors consistent with their established role in cell-volume control. WNK kinases may also be regulated by hydrostatic pressure. Hydrostatic pressure applied to cells in culture with N2 gas or to Drosophila Malpighian tubules by centrifugation induces phosphorylation of downstream effectors of endogenous WNKs. In vitro, the autophosphorylation and activity of the unphosphorylated kinase domain of WNK3 (uWNK3) is enhanced to a lesser extent than in cells by 190 kPa applied with N2 gas. Hydrostatic pressure measurably alters the structure of uWNK3. Data from size exclusion chromatography in line with multi-angle light scattering (SEC-MALS), SEC alone at different back pressures, analytical ultracentrifugation (AUC), NMR, and chemical crosslinking indicate a change in oligomeric structure in the presence of hydrostatic pressure from a WNK3 dimer to a monomer. The effects on the structure are related to those seen with osmolytes. Potential mechanisms of hydrostatic pressure activation of uWNK3 and the relationships of pressure activation to WNK osmosensing are discussed.
Collapse
Affiliation(s)
- John M. Humphreys
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Liliana R. Teixeira
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Radha Akella
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Haixia He
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ashari R. Kannangara
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kamil Sekulski
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John Pleinis
- Department of Internal Medicine, Division of Nephrology and Hypertension and Department of Human Genetics, University of Utah, Salt Lake City UT 84112
| | - Joanna Liwocha
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jenny Jiou
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kelly A. Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lukasz Joachimiak
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Josep Rizo
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Melanie H. Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Chad A. Brautigam
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Aylin R. Rodan
- Department of Internal Medicine, Division of Nephrology and Hypertension and Department of Human Genetics, University of Utah, Salt Lake City UT 84112
| | - Elizabeth J. Goldsmith
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
8
|
Liu K, Liu J, Liu Y, Wang H, Wang Z, Liu J, Wen S. Association study of WNK1 genetic variants and essential hypertension risk in the Northern Han Chinese in Beijing. Front Genet 2023; 14:1234536. [PMID: 37779914 PMCID: PMC10541150 DOI: 10.3389/fgene.2023.1234536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023] Open
Abstract
Background: Essential hypertension (EH) is a complex disorder resulting from interaction of genetic and environmental factors. Lysine deficient protein kinase 1 (WNK1) plays a very important role in maintaining renal potassium, sodium and chlorine ions balance as well as the regulation of blood pressure, so the WNK1 gene is considered a key gene for EH. This study thus sought to evaluate possible genetic associations between the WNK1 genetic variants and EH risk in the Northern Han Chinese population in Beijing. Methods: This study included 476 hypertensive subjects and 491 normotensive subjects. A total of 12 tag SNVs of WNK1 gene were genotyped successfully by TaqMan assay. Comparisons of the genotypic and allelic frequency between cases and controls were made by using the chi-square test. Logistic regression analyses were performed under different genetic models, and haplotype analysis was also conducted. Results: A total of 12 SNVs were identified as the tag SNVs for WNK1 gene. Significant associations were observed between WNK1 gene rs7305099 variant and EH risk, and T allele influenced hypertension risk in a protective manner. After correcting for multiple testing using Bonferroni, the significance remained for the SNV of rs7305099 in three genetic models [allele comparison, p < 0.0002, OR = 0.627, 95%CI (0.491-0.801); homozygote comparison, p < 0.0003, OR = 0.278, 95%CI (0.140-0.552); additive model, p < 0.0003, OR = 0.279, 95%CI (0.140-0.553)]. In the haplotype analyses, we found that the haplotype A-A-A-C-G-G-G was significantly associated with increased risk for EH (p = 0.043, OR = 1.23). Conclusion: Our data suggested that the rs7305099 genetic variant and the haplotype A-A-A-C-G-G-G on WNK1 gene might be associated with the susceptibility of EH in the Northern Han Chinese population. These could provide evidences to the risk assessment, early prevention and individualized therapy of EH to some extent.
Collapse
Affiliation(s)
- Kuo Liu
- Department of Hypertension Research, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Jielin Liu
- Department of Hypertension Research, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Ya Liu
- Department of Hypertension Research, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Hao Wang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Zuoguang Wang
- Department of Hypertension Research, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Jinghua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Shaojun Wen
- Department of Hypertension Research, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Loeillet S, Nicolas A. DNA polymerase δ: A single Pol31 polymorphism suppresses the strain background-specific lethality of Pol32 inactivation in Saccharomyces cerevisiae. DNA Repair (Amst) 2023; 127:103514. [PMID: 37244009 DOI: 10.1016/j.dnarep.2023.103514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/29/2023]
Abstract
The evolutionarily conserved DNA polymerase delta (Polδ) plays several essential roles in eukaryotic DNA replication and repair, responsible for the synthesis of the lagging-strand, lower replicative mutagenesis via its proof-reading exonuclease activity and synthetizes both strands during break-induced replication. In Saccharomyces cerevisiae, the Polδ protein complex consists of three subunits encoded by the POL3, POL31 and POL32 genes. Surprisingly, in contrast to POL3 and POL31, the POL32 gene deletion was found to be viable but lethal in all other eukaryotes, raising the question to which extent the viability of the POL32 deletion in S. cerevisiae was species specific. To address this issue, we inactivated the POL32 gene in 10 evolutionary close or distant S. cerevisiae strains and found that POL32 was either essential (3 strains including SK1), non-essential (5 strains including the reference S288C strain) or confers a slow-growth phenotype (2 strains). Whole-genome sequencing of S288C/SK1 pol32∆ meiotic segregants identified the lethal/suppressor effect of the single Pol31-C43Y polymorphism. Consistently, the introduction of the Pol31-43C allele in the SK1 and West African (WA) pol32∆ mutants was sufficient to restore cell viability and wild-type growth upon introduction of two copies of POL31-43C in the SK1 haploid strain. Reciprocally, introduction of the SK1 POL31-43Y allele in the S288C pol32∆ mutant was lethal. Sequence analyses of the POL31 polymorphisms in the 1,011 yeasts genome dataset correlates with the strict occurrence of the POL31-43Y allele in the yeast African palm wine clade. Differently, the single Pol31-E400G polymorphism confers pol32∆ lethality in the Malaysian strain. In the yeast two-hybrid assay, we observed a weakened interaction between Pol3 and Pol31-43Y versus Pol31-43C suggesting an insufficient level of the Polδ holoenzyme stability/activity. Thus, the enigmatic non-essentiality of Pol32 in S. cerevisiae results from single Pol31 amino acid polymorphism and is clade rather than species specific.
Collapse
Affiliation(s)
- S Loeillet
- Institut Curie Research Center, CNRS UMR3244, PSL Research University, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - A Nicolas
- Institut Curie Research Center, CNRS UMR3244, PSL Research University, 26 rue d'Ulm, 75248 Paris Cedex 05, France; IRCAN, CNRS UMR7284, INSERM U1081, Université Côte d'Azur, 28 avenue de Valombrose, 06107 Nice, France.
| |
Collapse
|
10
|
Hayward DA, Vanes L, Wissmann S, Sivapatham S, Hartweger H, Biggs O’May J, de Boer LL, Mitter R, Köchl R, Stein JV, Tybulewicz VL. B cell-intrinsic requirement for WNK1 kinase in antibody responses in mice. J Exp Med 2023; 220:e20211827. [PMID: 36662229 PMCID: PMC9872328 DOI: 10.1084/jem.20211827] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/20/2022] [Accepted: 12/23/2022] [Indexed: 01/21/2023] Open
Abstract
Migration and adhesion play critical roles in B cells, regulating recirculation between lymphoid organs, migration within lymphoid tissue, and interaction with CD4+ T cells. However, there is limited knowledge of how B cells integrate chemokine receptor and integrin signaling with B cell activation to generate efficient humoral responses. Here, we show that the WNK1 kinase, a regulator of migration and adhesion, is essential in B cells for T-dependent and -independent antibody responses. We demonstrate that WNK1 transduces signals from the BCR, CXCR5, and CD40, and using intravital imaging, we show that WNK1 regulates migration of naive and activated B cells, and their interactions with T cells. Unexpectedly, we show that WNK1 is required for BCR- and CD40-induced proliferation, acting through the OXSR1 and STK39 kinases, and for efficient B cell-T cell collaboration in vivo. Thus, WNK1 is critical for humoral immune responses, by regulating B cell migration, adhesion, and T cell-dependent activation.
Collapse
Affiliation(s)
| | | | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Sujana Sivapatham
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | | | | | | | | | | | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
11
|
Chlebowicz J, Akella R, Humphreys JM, He H, Kannangara AR, Wei S, Posner B, Goldsmith EJ. Identification of a Class of WNK Isoform-Specific Inhibitors Through High-Throughput Screening. Drug Des Devel Ther 2023; 17:93-105. [PMID: 36712947 PMCID: PMC9880028 DOI: 10.2147/dddt.s389461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/17/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction WNK [with no lysine (K)] kinases are serine/threonine kinases associated with familial hyperkalemic hypertension (FHHt). WNKs are therapeutic targets for blood pressure regulation, stroke and several cancers including triple negative breast cancer and glioblastoma. Here, we searched for and characterized novel WNK kinase inhibitors. Methods We used a ~210,000-compound library in a high-throughput screen, re-acquisition and assay, commercial specificity screens and crystallography to identify WNK-isoform-selective inhibitors. Results We identified five classes of compounds that inhibit the kinase activity of WNK1: quinoline compounds, halo-sulfones, cyclopropane-containing thiazoles, piperazine-containing compounds, and nitrophenol-derived compounds. The compounds are strongly pan-WNK selective, inhibiting all four WNK isoforms. A class of quinoline compounds was identified that further shows selectivity among the WNK isoforms, being more potent toward WNK3 than WNK1. The crystal structure of the quinoline-derived SW120619 bound to the kinase domain of WNK3 reveals active site binding, and comparison to the WNK1 structure reveals the potential origin of isoform specificity. Discussion The newly discovered classes of compounds may be starting points for generating pharmacological tools and potential drugs treating hypertension and cancer.
Collapse
Affiliation(s)
- Julita Chlebowicz
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Radha Akella
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John M Humphreys
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haixia He
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ashari R Kannangara
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shuguang Wei
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce Posner
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth J Goldsmith
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA,Correspondence: Elizabeth J Goldsmith, Department of Biophysics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8816, USA, Tel +1 214 645 6376, Email
| |
Collapse
|
12
|
Yoon HJ, Kim GC, Oh S, Kim H, Kim YK, Lee Y, Kim MS, Kwon G, Ok YS, Kwon HK, Kim HS. WNK3 inhibition elicits antitumor immunity by suppressing PD-L1 expression on tumor cells and activating T-cell function. Exp Mol Med 2022; 54:1913-1926. [PMID: 36357569 PMCID: PMC9722663 DOI: 10.1038/s12276-022-00876-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/24/2022] [Accepted: 08/17/2022] [Indexed: 11/12/2022] Open
Abstract
Immune checkpoint therapies, such as programmed cell death ligand 1 (PD-L1) blockade, have shown remarkable clinical benefit in many cancers by restoring the function of exhausted T cells. Hence, the identification of novel PD-L1 regulators and the development of their inhibition strategies have significant therapeutic advantages. Here, we conducted pooled shRNA screening to identify regulators of membrane PD-L1 levels in lung cancer cells targeting druggable genes and cancer drivers. We identified WNK lysine deficient protein kinase 3 (WNK3) as a novel positive regulator of PD-L1 expression. The kinase-dead WNK3 mutant failed to elevate PD-L1 levels, indicating the involvement of its kinase domain in this function. WNK3 perturbation increased cancer cell death in cancer cell-immune cell coculture conditions and boosted the secretion of cytokines and cytolytic enzymes, promoting antitumor activities in CD4+ and CD8+ T cells. WNK463, a pan-WNK inhibitor, enhanced CD8+ T-cell-mediated antitumor activity and suppressed tumor growth as a monotherapy as well as in combination with a low-dose anti-PD-1 antibody in the MC38 syngeneic mouse model. Furthermore, we demonstrated that the c-JUN N-terminal kinase (JNK)/c-JUN pathway underlies WNK3-mediated transcriptional regulation of PD-L1. Our findings highlight that WNK3 inhibition might serve as a potential therapeutic strategy for cancer immunotherapy through its concurrent impact on cancer cells and immune cells.
Collapse
Affiliation(s)
- Hyun Ju Yoon
- grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Gi-Cheon Kim
- grid.15444.300000 0004 0470 5454Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sejin Oh
- grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Hakhyun Kim
- grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Keon Kim
- grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Yunji Lee
- grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Min Seo Kim
- grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Gino Kwon
- grid.15444.300000 0004 0470 5454Graduate Program for Nanomedical Science, Yonsei University, Seoul, Korea
| | - Yeon-Su Ok
- grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
| | - Ho-Keun Kwon
- grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Seok Kim
- grid.15444.300000 0004 0470 5454Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea ,grid.15444.300000 0004 0470 5454Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Rodriguez M, Kannangara A, Chlebowicz J, Akella R, He H, Tambar UK, Goldsmith EJ. Synthesis and Structural Characterization of Novel Trihalo-sulfone Inhibitors of WNK1. ACS Med Chem Lett 2022; 13:1678-1684. [PMID: 36262391 PMCID: PMC9575160 DOI: 10.1021/acsmedchemlett.2c00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022] Open
Abstract
With No lysine (K) [WNK] kinases are structurally unique serine/threonine protein kinases that have therapeutic potential for blood pressure regulation and cancer. A novel class of trihalo-sulfone compounds was identified by high-throughput screening. Trihalo-sulfone 1 emerged as an effective inhibitor of WNK1 with an IC50 value of 1.6 μM. Herein, we define chemical features necessary for inhibition of WNK1 using chemical synthesis and X-ray crystallography. Analogues that probed the role of specific functional groups to the inhibitory activity were synthesized. X-ray structures of trihalo-sulfone 1 and a second trihalo-sulfone 23 bound to WNK1 revealed active site binding to two of the three previously defined canonical inhibitor binding pockets as well as a novel binding site for the trihalo-sulfone moiety. The elucidation of these novel interaction sites may allow for the strategic design of even more selective and potent WNK inhibitors.
Collapse
Affiliation(s)
- Melanie Rodriguez
- Department
of Biochemistry, The University of Texas
Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| | - Ashari Kannangara
- Department
of Biophysics, The University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8816, United States
| | - Julita Chlebowicz
- Department
of Biophysics, The University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8816, United States
| | - Radha Akella
- Department
of Biophysics, The University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8816, United States
| | - Haixia He
- Department
of Biophysics, The University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8816, United States
| | - Uttam K. Tambar
- Department
of Biochemistry, The University of Texas
Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| | - Elizabeth J. Goldsmith
- Department
of Biophysics, The University of Texas Southwestern
Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8816, United States
| |
Collapse
|
14
|
Rautureau Y, Berlatie M, Rivas D, Uy K, Blanchette A, Miquel G, Higgins MÈ, Mecteau M, Nault A, Villeneuve L, Lavoie V, Théberge-Julien G, Brand G, Lapointe L, Denis M, Rosa C, Fortier A, Blondeau L, Guertin MC, Dubé MP, Thorin É, Ledoux J, Rhainds D, Rhéaume É, Tardif JC. Adenylate cyclase type 9 antagonizes cAMP accumulation and regulates endothelial signaling involved in atheroprotection. Cardiovasc Res 2022; 119:450-464. [PMID: 35576489 DOI: 10.1093/cvr/cvac085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS The adenylate cyclase type 9 (ADCY9) gene appears to determine atherosclerotic outcomes in patients treated with dalcetrapib. In mice, we recently demonstrated that Adcy9 inactivation potentiates endothelial function and inhibits atherogenesis. The objective of this study was to characterize the contribution of ADCY9 to the regulation of endothelial signaling pathways involved in atherosclerosis. METHODS AND RESULTS We show that ADCY9 is expressed in the endothelium of mouse aorta and femoral arteries. We demonstrate that ADCY9 inactivation in cultured endothelial cells paradoxically increases cAMP accumulation in response to the adenylate cyclase activators forskolin and vasoactive intestinal peptide (VIP). Reciprocally, ADCY9 overexpression decreases cAMP production. Using mouse femoral artery arteriography, we show that Adcy9 inactivation potentiates VIP-induced endothelial-dependent vasodilation. Moreover, Adcy9 inactivation reduces mouse atheroma endothelial permeability in different vascular beds. ADCY9 overexpression reduces forskolin-induced phosphorylation of Ser157-vasodilator-stimulated phosphoprotein (VASP) and worsens thrombin-induced fall of RAP1 activity, both leading to increased endothelial permeability. ADCY9 inactivation in thrombin-stimulated human coronary artery endothelial cells results in cAMP accumulation, increases p-Ser157-VASP and inhibits endothelial permeability. MLC2 phosphorylation and actin stress fiber increases in response to thrombin were reduced by ADCY9 inactivation, suggesting actin cystoskeleton regulation. Finally, using the Miles assay, we demonstrate that Adcy9 regulates thrombin-induced endothelial permeability in vivo in normal and atherosclerotic animals. CONCLUSION Adcy9 is expressed in endothelial cells and regulates local cAMP and endothelial functions including permeability relevant to atherogenesis.
Collapse
Affiliation(s)
- Yohann Rautureau
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | | | - Daniel Rivas
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Kurunradeth Uy
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Alexandre Blanchette
- Department of Physiology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Géraldine Miquel
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | | | - Mélanie Mecteau
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Audrey Nault
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Véronique Lavoie
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | | | - Geneviève Brand
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Line Lapointe
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Maxime Denis
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Camille Rosa
- Montreal Health Innovations Coordinating Centre (MHICC), Montreal, Canada
| | - Annik Fortier
- Montreal Health Innovations Coordinating Centre (MHICC), Montreal, Canada
| | - Lucie Blondeau
- Montreal Health Innovations Coordinating Centre (MHICC), Montreal, Canada
| | | | - Marie-Pierre Dubé
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada.,Department of Medicine
| | - Éric Thorin
- Montreal Heart Institute, Université de Montréal, Montreal, Canada.,Department of Surgery of the Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Jonathan Ledoux
- Department of Physiology and Pharmacology, Université de Montréal, Montreal, Canada
| | - David Rhainds
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Éric Rhéaume
- Montreal Heart Institute, Université de Montréal, Montreal, Canada.,Department of Medicine
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, Montreal, Canada.,Department of Medicine
| |
Collapse
|
15
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
16
|
Hou CY, Ma CY, Yuh CH. WNK1 kinase signaling in metastasis and angiogenesis. Cell Signal 2022; 96:110371. [DOI: 10.1016/j.cellsig.2022.110371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023]
|
17
|
Powell DR, Doree DD, DaCosta CM, Platt KA, Brommage R, Buhring L, Revelli JP, Shadoan MK. Mice Lacking Gpr75 are Hypophagic and Thin. Diabetes Metab Syndr Obes 2022; 15:45-58. [PMID: 35023939 PMCID: PMC8743382 DOI: 10.2147/dmso.s342799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/15/2021] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Humans with haploinsufficiency of GPR75, an orphan GPCR, are thin. Gpr75 knockout (KO) mice are also thin with improved glucose homeostasis. We wanted to confirm these findings in Gpr75 KO mice and determine whether decreased energy intake and/or increased energy expenditure contributed to the thin phenotype. METHODS Gpr75 KO mice were generated by homologous recombination. All studies compared female and male Gpr75 KO mice to their wild type (WT) littermates. Body composition was measured by DXA and QMR technologies. Glucose homeostasis was evaluated by measuring glucose and insulin levels during oral glucose tolerance tests (OGTTs). Food intake was measured in group-housed mice. In singly housed mice, energy expenditure was measured in Oxymax indirect calorimetry chambers, and locomotor activity was measured in Oxymax and Photobeam Activity System chambers. RESULTS In all 12 cohorts of adult female or male mice, Gpr75 KO mice had less body fat; pooled data showed that, compared to WT littermates (n = 103), Gpr75 KO mice (n = 118) had 49% less body fat and 4% less LBM (P < 0.001 for each). KO mice also had 8% less body fat at weaning (P < 0.05), and during the month after weaning as the thin phenotype became more exaggerated, Gpr75 KO mice ate significantly less than, but had energy expenditure and activity levels comparable to, their WT littermates. During OGTTs, Gpr75 KO mice showed improved glucose tolerance (glucose AUC 23% lower in females, P < 0.05, and 26% lower in males, P < 0.001), accompanied by significantly decreased insulin levels and significantly increased insulin sensitivity indices. CONCLUSION Gpr75 KO mice are thin at weaning, are hypophagic as the thin phenotype becomes more exaggerated, and exhibit improved glucose tolerance and insulin sensitivity as healthy-appearing adults. These results suggest that inhibiting GPR75 in obese humans may safely decrease energy intake and body fat while improving glucose tolerance and insulin sensitivity.
Collapse
Affiliation(s)
- David R Powell
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Correspondence: David R Powell Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc., 2445 Technology Forest Boulevard, The Woodlands, TX, 77381, USATel +1 713 249 3972Fax +1 281 863 8115 Email
| | - Deon D Doree
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Christopher M DaCosta
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Kenneth A Platt
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc., The Woodlands, TX, USA
| | - Robert Brommage
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Lindsey Buhring
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Jean-Pierre Revelli
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Melanie K Shadoan
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| |
Collapse
|
18
|
Lim WM, Chin EWM, Tang BL, Chen T, Goh ELK. WNK3 Maintains the GABAergic Inhibitory Tone, Synaptic Excitation and Neuronal Excitability via Regulation of KCC2 Cotransporter in Mature Neurons. Front Mol Neurosci 2021; 14:762142. [PMID: 34858138 PMCID: PMC8631424 DOI: 10.3389/fnmol.2021.762142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
The activation of chloride (Cl−)permeable gamma (γ)-aminobutyric acid type A(GABAA) receptors induces synaptic inhibition in mature and excitation in immature neurons. This developmental “switch” in GABA function controlled by its polarity depends on the postnatal decrease in intraneuronal Cl− concentration mediated by KCC2, a member of cation-chloride cotransporters (CCCs). The serine-threonine kinase WNK3 (With No Lysine [K]), is a potent regulator of all CCCs and is expressed in neurons. Here, we characterized the functions of WNK3 and its role in GABAergic signaling in cultured embryonic day 18 (E18) hippocampal neurons. We observed a decrease in WNK3 expression as neurons mature. Knocking down of WNK3 significantly hyperpolarized EGABA in mature neurons (DIV13–15) but had no effect on immature neurons (DIV6–8). This hyperpolarized EGABA in WNK3-deficient neurons was not due to the total expression of NKCC1 and KCC2, that remained unchanged. However, there was a reduction in phosphorylated KCC2 at the membrane, suggesting an increase in KCC2 chloride export activity. Furthermore, hyperpolarized EGABA observed in WNK3-deficient neurons can be reversed by the KCC2 inhibitor, VU024055, thus indicating that WNK3 acts through KCC2 to influence EGABA. Notably, WNK3 knockdown resulted in morphological changes in mature but not immature neurons. Electrophysiological characterization of WNK3-deficient mature neurons revealed reduced capacitances but increased intrinsic excitability and synaptic excitation. Hence, our study demonstrates that WNK3 maintains the “adult” GABAergic inhibitory tone in neurons and plays a role in the morphological development of neurons and excitability.
Collapse
Affiliation(s)
- Wee Meng Lim
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore
| | - Eunice W M Chin
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bor Luen Tang
- NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tingting Chen
- School of Pharmacy, Nantong University, Nantong, China
| | - Eyleen L K Goh
- Neuroscience Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
19
|
Vogel P, Read RW, Hansen GM, Powell DR. Histopathology is required to identify and characterize myopathies in high-throughput phenotype screening of genetically engineered mice. Vet Pathol 2021; 58:1158-1171. [PMID: 34269122 DOI: 10.1177/03009858211030541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The development of mouse models that replicate the genetic and pathological features of human disease is important in preclinical research because these types of models enable the completion of meaningful pharmacokinetic, safety, and efficacy studies. Numerous relevant mouse models of human disease have been discovered in high-throughput screening programs, but there are important specific phenotypes revealed by histopathology that are not reliably detected by any other physiological or behavioral screening tests. As part of comprehensive phenotypic analyses of over 4000 knockout (KO) mice, histopathology identified 12 lines of KO mice with lesions indicative of an autosomal recessive myopathy. This report includes a brief summary of histological and other findings in these 12 lines. Notably, the inverted screen test detected muscle weakness in only 4 of these 12 lines (Scyl1, Plpp7, Chkb, and Asnsd1), all 4 of which have been previously recognized and published. In contrast, 6 of 8 KO lines showing negative or inconclusive findings on the inverted screen test (Plppr2, Pnpla7, Tenm1, Srpk3, Sidt2, Yif1b, Mrs2, and Pnpla2) had not been previously identified as having myopathies. These findings support the need to include histopathology in phenotype screening protocols in order to identify novel genetic myopathies that are not clinically evident or not detected by the inverted screen test.
Collapse
Affiliation(s)
- Peter Vogel
- 5417St Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert W Read
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | | | - David R Powell
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| |
Collapse
|
20
|
Powell DR, Revelli JP, Doree DD, DaCosta CM, Desai U, Shadoan MK, Rodriguez L, Mullens M, Yang QM, Ding ZM, Kirkpatrick LL, Vogel P, Zambrowicz B, Sands AT, Platt KA, Hansen GM, Brommage R. High-Throughput Screening of Mouse Gene Knockouts Identifies Established and Novel High Body Fat Phenotypes. Diabetes Metab Syndr Obes 2021; 14:3753-3785. [PMID: 34483672 PMCID: PMC8409770 DOI: 10.2147/dmso.s322083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/04/2021] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Obesity is a major public health problem. Understanding which genes contribute to obesity may better predict individual risk and allow development of new therapies. Because obesity of a mouse gene knockout (KO) line predicts an association of the orthologous human gene with obesity, we reviewed data from the Lexicon Genome5000TM high throughput phenotypic screen (HTS) of mouse gene KOs to identify KO lines with high body fat. MATERIALS AND METHODS KO lines were generated using homologous recombination or gene trapping technologies. HTS body composition analyses were performed on adult wild-type and homozygous KO littermate mice from 3758 druggable mouse genes having a human ortholog. Body composition was measured by either DXA or QMR on chow-fed cohorts from all 3758 KO lines and was measured by QMR on independent high fat diet-fed cohorts from 2488 of these KO lines. Where possible, comparisons were made to HTS data from the International Mouse Phenotyping Consortium (IMPC). RESULTS Body fat data are presented for 75 KO lines. Of 46 KO lines where independent external published and/or IMPC KO lines are reported as obese, 43 had increased body fat. For the remaining 29 novel high body fat KO lines, Ksr2 and G2e3 are supported by data from additional independent KO cohorts, 6 (Asnsd1, Srpk2, Dpp8, Cxxc4, Tenm3 and Kiss1) are supported by data from additional internal cohorts, and the remaining 21 including Tle4, Ak5, Ntm, Tusc3, Ankk1, Mfap3l, Prok2 and Prokr2 were studied with HTS cohorts only. CONCLUSION These data support the finding of high body fat in 43 independent external published and/or IMPC KO lines. A novel obese phenotype was identified in 29 additional KO lines, with 27 still lacking the external confirmation now provided for Ksr2 and G2e3 KO mice. Undoubtedly, many mammalian obesity genes remain to be identified and characterized.
Collapse
Affiliation(s)
- David R Powell
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Jean-Pierre Revelli
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Deon D Doree
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Christopher M DaCosta
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Urvi Desai
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Melanie K Shadoan
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Lawrence Rodriguez
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Michael Mullens
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Qi M Yang
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Zhi-Ming Ding
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Laura L Kirkpatrick
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Peter Vogel
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| | - Brian Zambrowicz
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Arthur T Sands
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
- Department of Information Technology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Kenneth A Platt
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Gwenn M Hansen
- Department of Molecular Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, Tx, USA
| | - Robert Brommage
- Department of Pharmaceutical Biology, Lexicon Pharmaceuticals, Inc, The Woodlands, TX, USA
| |
Collapse
|
21
|
Mayes-Hopfinger L, Enache A, Xie J, Huang CL, Köchl R, Tybulewicz VLJ, Fernandes-Alnemri T, Alnemri ES. Chloride sensing by WNK1 regulates NLRP3 inflammasome activation and pyroptosis. Nat Commun 2021; 12:4546. [PMID: 34315884 PMCID: PMC8316491 DOI: 10.1038/s41467-021-24784-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/30/2021] [Indexed: 01/05/2023] Open
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.
Collapse
Affiliation(s)
- Lindsey Mayes-Hopfinger
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aura Enache
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jian Xie
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Chou-Long Huang
- Department of Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Robert Köchl
- The Francis Crick Institute, London, UK
- Kings College London, London, UK
| | | | - Teresa Fernandes-Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Taylor CA, Cobb MH. CCT and CCT-like Modular Protein Interaction Domains in WNK Signaling. Mol Pharmacol 2021; 101:201-212. [PMID: 34312216 DOI: 10.1124/molpharm.121.000307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022] Open
Abstract
The WNK (with-no lysine (K)) kinases and their downstream effector kinases, OSR1 (oxidative stress responsive 1) and SPAK (SPS/STE20-related proline-alanine rich kinase), have well-established functions in the maintenance of cell volume and ion homeostasis. Mutations in these kinases have been linked to an inherited form of hypertension, neurological defects, and other pathologies. A rapidly expanding body of evidence points to the involvement of WNKs in regulating multiple diverse cellular processes as well as the progression of some forms of cancer. How OSR1/SPAK contribute to these processes is well understood in some cases, but completely unknown in others. OSR1 and SPAK are targeted to both WNKs and substrates via their conserved C-terminal (CCT) protein interaction domains. Considerable effort has been put forth to understand the structure, function, and interaction specificity of the CCT domains in relation to WNK signaling, and multiple inhibitors of WNK signaling target these domains. The domains bind RFxV and RxFxV protein sequence motifs with the consensus sequence R-F-x-V/I or R-x-F-x-V/I, but residues outside the core motif also contribute to specificity. CCT interactions are required for OSR1 and SPAK activation and deactivation as well as cation-chloride cotransporter substrate phosphorylation. All four WNKs also contain CCT-like domains that have similar structures and conserved binding residues when compared to CCT domains, but their functions and interaction specificities are mostly unknown. A better understanding of the varied actions of these domains and their interactions will better define the known signaling mechanisms of the WNK pathway as well as uncover new ones. Significance Statement WNK kinases and downstream effector kinases, OSR1 and SPAK, have been shown to be involved in an array of diverse cellular processes. Here we review the function of modular protein interaction domains found in OSR1 and SPAK as well as related domains found in WNKs.
Collapse
Affiliation(s)
- Clinton A Taylor
- Pharmacology, University of Texas Southwestern Medical Center, United States
| | - Melanie H Cobb
- Pharmacology, University of Texas Southwestern Medical Center, United States
| |
Collapse
|
23
|
Jaykumar AB, Jung JU, Parida PK, Dang TT, Wichaidit C, Kannangara AR, Earnest S, Goldsmith EJ, Pearson GW, Malladi S, Cobb MH. WNK1 Enhances Migration and Invasion in Breast Cancer Models. Mol Cancer Ther 2021; 20:1800-1808. [PMID: 34253593 DOI: 10.1158/1535-7163.mct-21-0174] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/28/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022]
Abstract
Metastasis is the major cause of mortality in patients with breast cancer. Many signaling pathways have been linked to cancer invasiveness, but blockade of few protein components has succeeded in reducing metastasis. Thus, identification of proteins contributing to invasion that are manipulable by small molecules may be valuable in inhibiting spread of the disease. The protein kinase with no lysine (K) 1 (WNK1) has been suggested to induce migration of cells representing a range of cancer types. Analyses of mouse models and patient data have implicated WNK1 as one of a handful of genes uniquely linked to invasive breast cancer. Here, we present evidence that inhibition of WNK1 slows breast cancer metastasis. We show that depletion or inhibition of WNK1 reduces migration of several breast cancer cell lines in wound healing assays and decreases invasion in collagen matrices. Furthermore, WNK1 depletion suppresses expression of AXL, a tyrosine kinase implicated in metastasis. Finally, we demonstrate that WNK inhibition in mice attenuates tumor progression and metastatic burden. These data showing reduced migration, invasion, and metastasis upon WNK1 depletion in multiple breast cancer models suggest that WNK1 contributes to the metastatic phenotype, and that WNK1 inhibition may offer a therapeutic avenue for attenuating progression of invasive breast cancers.
Collapse
Affiliation(s)
- Ankita B Jaykumar
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| | - Ji-Ung Jung
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| | | | - Tuyen T Dang
- Department of Molecular Oncology, Georgetown University, Washington, District of Columbia
| | | | | | - Svetlana Earnest
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| | | | - Gray W Pearson
- Department of Molecular Oncology, Georgetown University, Washington, District of Columbia
| | - Srinivas Malladi
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Melanie H Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
24
|
Brown A, Meor Azlan NF, Wu Z, Zhang J. WNK-SPAK/OSR1-NCC kinase signaling pathway as a novel target for the treatment of salt-sensitive hypertension. Acta Pharmacol Sin 2021; 42:508-517. [PMID: 32724175 PMCID: PMC8115323 DOI: 10.1038/s41401-020-0474-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/06/2020] [Indexed: 02/08/2023] Open
Abstract
Hypertension is the most prevalent health condition worldwide, affecting ~1 billion people. Gordon's syndrome is a form of secondary hypertension that can arise due to a number of possible mutations in key genes that encode proteins in a pathway containing the With No Lysine [K] (WNK) and its downstream target kinases, SPS/Ste20-related proline-alanine-rich kinase (SPAK) and oxidative stress responsive kinase 1 (OSR1). This pathway regulates the activity of the thiazide-sensitive sodium chloride cotransporter (NCC), which is responsible for NaCl reabsorption in the distal nephron. Therefore, mutations in genes encoding proteins that regulate the NCC proteins disrupt ion homeostasis and cause hypertension by increasing NaCl reabsorption. Thiazide diuretics are currently the main treatment option for Gordon's syndrome. However, they have a number of side effects, and chronic usage can lead to compensatory adaptations in the nephron that counteract their action. Therefore, recent research has focused on developing novel inhibitory molecules that inhibit components of the WNK-SPAK/OSR1-NCC pathway, thereby reducing NaCl reabsorption and restoring normal blood pressure. In this review we provide an overview of the currently reported molecular inhibitors of the WNK-SPAK/OSR1-NCC pathway and discuss their potential as treatment options for Gordon's syndrome.
Collapse
Affiliation(s)
- Archie Brown
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Zhijuan Wu
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
- Newcastle University Business School, Newcastle University, Newcastle upon Tyne, NE1 4SE, UK
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK.
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361004, China.
| |
Collapse
|
25
|
Control of Protein and Energy Metabolism in the Pituitary Gland in Response to Three-Week Running Training in Adult Male Mice. Cells 2021; 10:cells10040736. [PMID: 33810540 PMCID: PMC8065971 DOI: 10.3390/cells10040736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
It is assumed that crosstalk of central and peripheral tissues plays a role in the adaptive response to physical activity and exercise. Here, we wanted to study the effects of training and genetic predisposition in a marathon mouse model on mRNA expression in the pituitary gland. Therefore, we used a mouse model developed by phenotype selection for superior running performance (DUhTP) and non-inbred control mice (DUC). Both mouse lines underwent treadmill training for three weeks or were kept in a sedentary condition. In all groups, total RNA was isolated from the pituitary gland and sequenced. Molecular pathway analysis was performed by ingenuity pathway analysis (IPA). Training induced differential expression of 637 genes (DEGs) in DUC but only 50 DEGs in DUhTP mice. Genetic selection for enhanced running performance strongly affected gene expression in the pituitary gland and identified 1732 DEGs in sedentary DUC versus DUhTP mice. Training appeared to have an even stronger effect on gene expression in both lines and comparatively revealed 3828 DEGs in the pituitary gland. From the list of DEGs in all experimental groups, candidate genes were extracted by comparison with published genomic regions with significant effects on training responses in mice. Bioinformatic modeling revealed induction and coordinated expression of the pathways for ribosome synthesis and oxidative phosphorylation in DUC mice. By contrast, DUhTP mice were resistant to the positive effects of three-week training on protein and energy metabolism in the pituitary gland.
Collapse
|
26
|
Gomi H, Hinata A, Yasui T, Torii S, Hosaka M. Expression Pattern of the LacZ Reporter in Secretogranin III Gene-trapped Mice. J Histochem Cytochem 2021; 69:229-243. [PMID: 33622062 DOI: 10.1369/0022155421996845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Secretogranin III (SgIII) is a granin protein involved in secretory granule formation in peptide-hormone-producing endocrine cells. In this study, we analyzed the expression of the LacZ reporter in the SgIII knockout mice produced by gene trapping (SgIII-gtKO) for the purpose of comprehensively clarifying the expression patterns of SgIII at the cell and tissue levels. In the endocrine tissues of SgIII-gtKO mice, LacZ expression was observed in the pituitary gland, adrenal medulla, and pancreatic islets, where SgIII expression has been canonically revealed. LacZ expression was extensively observed in brain regions, especially in the cerebral cortex, hippocampus, hypothalamic nuclei, cerebellum, and spinal cord. In peripheral nervous tissues, LacZ expression was observed in the retina, optic nerve, and trigeminal ganglion. LacZ expression was particularly prominent in astrocytes, in addition to neurons and ependymal cells. In the cerebellum, at least four cell types expressed SgIII under basal conditions. The expression of SgIII in the glioma cell lines C6 and RGC-6 was enhanced by excitatory glutamate treatment. It also became clear that the expression level of SgIII varied among neuron and astrocyte subtypes. These results suggest that SgIII is involved in glial cell function, in addition to neuroendocrine functions, in the nervous system.
Collapse
Affiliation(s)
- Hiroshi Gomi
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Airi Hinata
- Laboratory of Molecular Life Sciences, Department of Biotechnology, Akita Prefectural University, Akita, Japan
| | - Tadashi Yasui
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Seiji Torii
- Center for Food Science and Wellness, Gunma University, Maebashi, Japan
| | - Masahiro Hosaka
- Laboratory of Molecular Life Sciences, Department of Biotechnology, Akita Prefectural University, Akita, Japan
| |
Collapse
|
27
|
Hansen J, von Melchner H, Wurst W. Mutant non-coding RNA resource in mouse embryonic stem cells. Dis Model Mech 2021; 14:14/2/dmm047803. [PMID: 33729986 PMCID: PMC7875499 DOI: 10.1242/dmm.047803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/14/2020] [Indexed: 01/23/2023] Open
Abstract
Gene trapping is a high-throughput approach that has been used to introduce insertional mutations into the genome of mouse embryonic stem (ES) cells. It is performed with generic gene trap vectors that simultaneously mutate and report the expression of the endogenous gene at the site of insertion and provide a DNA sequence tag for the rapid identification of the disrupted gene. Large-scale international efforts assembled a gene trap library of 566,554 ES cell lines with single gene trap integrations distributed throughout the genome. Here, we re-investigated this unique library and identified mutations in 2202 non-coding RNA (ncRNA) genes, in addition to mutations in 12,078 distinct protein-coding genes. Moreover, we found certain types of gene trap vectors preferentially integrating into genes expressing specific long non-coding RNA (lncRNA) biotypes. Together with all other gene-trapped ES cell lines, lncRNA gene-trapped ES cell lines are readily available for functional in vitro and in vivo studies.
Collapse
Affiliation(s)
- Jens Hansen
- Institute of Developmental Genetics, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Harald von Melchner
- Department of Molecular Hematology, University Hospital Frankfurt, Goethe University, D-60590 Frankfurt am Main, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany .,Technische Universität München-Weihenstephan, c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Munich, Feodor-Lynen-Str. 17, D-81377 Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, D-81377 München, Germany
| |
Collapse
|
28
|
Kobaisi F, Fayyad N, Sulpice E, Badran B, Fayyad-Kazan H, Rachidi W, Gidrol X. High-throughput synthetic rescue for exhaustive characterization of suppressor mutations in human genes. Cell Mol Life Sci 2020; 77:4209-4222. [PMID: 32270227 PMCID: PMC7588364 DOI: 10.1007/s00018-020-03519-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023]
Abstract
Inherited or acquired mutations can lead to pathological outcomes. However, in a process defined as synthetic rescue, phenotypic outcome created by primary mutation is alleviated by suppressor mutations. An exhaustive characterization of these mutations in humans is extremely valuable to better comprehend why patients carrying the same detrimental mutation exhibit different pathological outcomes or different responses to treatment. Here, we first review all known suppressor mutations' mechanisms characterized by genetic screens on model species like yeast or flies. However, human suppressor mutations are scarce, despite some being discovered based on orthologue genes. Because of recent advances in high-throughput screening, developing an inventory of human suppressor mutations for pathological processes seems achievable. In addition, we review several screening methods for suppressor mutations in cultured human cells through knock-out, knock-down or random mutagenesis screens on large scale. We provide examples of studies published over the past years that opened new therapeutic avenues, particularly in oncology.
Collapse
Affiliation(s)
- Farah Kobaisi
- University of Grenoble Alpes, CEA, INSERM, IRIG-BGE U1038, 38000, Grenoble, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
- University of Grenoble Alpes, SYMMES/CIBEST UMR 5819 UGA-CNRS-CEA, IRIG/CEA-Grenoble, Grenoble, France
| | - Nour Fayyad
- University of Grenoble Alpes, SYMMES/CIBEST UMR 5819 UGA-CNRS-CEA, IRIG/CEA-Grenoble, Grenoble, France
| | - Eric Sulpice
- University of Grenoble Alpes, CEA, INSERM, IRIG-BGE U1038, 38000, Grenoble, France
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Walid Rachidi
- University of Grenoble Alpes, SYMMES/CIBEST UMR 5819 UGA-CNRS-CEA, IRIG/CEA-Grenoble, Grenoble, France
| | - Xavier Gidrol
- University of Grenoble Alpes, CEA, INSERM, IRIG-BGE U1038, 38000, Grenoble, France.
| |
Collapse
|
29
|
Murillo-de-Ozores AR, Chávez-Canales M, de los Heros P, Gamba G, Castañeda-Bueno M. Physiological Processes Modulated by the Chloride-Sensitive WNK-SPAK/OSR1 Kinase Signaling Pathway and the Cation-Coupled Chloride Cotransporters. Front Physiol 2020; 11:585907. [PMID: 33192599 PMCID: PMC7606576 DOI: 10.3389/fphys.2020.585907] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
The role of Cl- as an intracellular signaling ion has been increasingly recognized in recent years. One of the currently best described roles of Cl- in signaling is the modulation of the With-No-Lysine (K) (WNK) - STE20-Proline Alanine rich Kinase (SPAK)/Oxidative Stress Responsive Kinase 1 (OSR1) - Cation-Coupled Cl- Cotransporters (CCCs) cascade. Binding of a Cl- anion to the active site of WNK kinases directly modulates their activity, promoting their inhibition. WNK activation due to Cl- release from the binding site leads to phosphorylation and activation of SPAK/OSR1, which in turn phosphorylate the CCCs. Phosphorylation by WNKs-SPAK/OSR1 of the Na+-driven CCCs (mediating ions influx) promote their activation, whereas that of the K+-driven CCCs (mediating ions efflux) promote their inhibition. This results in net Cl- influx and feedback inhibition of WNK kinases. A wide variety of alterations to this pathway have been recognized as the cause of several human diseases, with manifestations in different systems. The understanding of WNK kinases as Cl- sensitive proteins has allowed us to better understand the mechanistic details of regulatory processes involved in diverse physiological phenomena that are reviewed here. These include cell volume regulation, potassium sensing and intracellular signaling in the renal distal convoluted tubule, and regulation of the neuronal response to the neurotransmitter GABA.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paola de los Heros
- Unidad de Investigación UNAM-INC, Research Division, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
30
|
Meor Azlan NF, Zhang J. Role of the Cation-Chloride-Cotransporters in Cardiovascular Disease. Cells 2020; 9:E2293. [PMID: 33066544 PMCID: PMC7602155 DOI: 10.3390/cells9102293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 02/05/2023] Open
Abstract
The SLC12 family of cation-chloride-cotransporters (CCCs) is comprised of potassium chloride cotransporters (KCCs), which mediate Cl- extrusion and sodium-potassium chloride cotransporters (N[K]CCs), which mediate Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. The functions of CCCs influence a variety of physiological processes, many of which overlap with the pathophysiology of cardiovascular disease. Although not all of the cotransporters have been linked to Mendelian genetic disorders, recent studies have provided new insights into their functional role in vascular and renal cells in addition to their contribution to cardiovascular diseases. Particularly, an imbalance in potassium levels promotes the pathogenesis of atherosclerosis and disturbances in sodium homeostasis are one of the causes of hypertension. Recent findings suggest hypothalamic signaling as a key signaling pathway in the pathophysiology of hypertension. In this review, we summarize and discuss the role of CCCs in cardiovascular disease with particular emphasis on knowledge gained in recent years on NKCCs and KCCs.
Collapse
Affiliation(s)
- Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, Hatherly Laboratories, University of Exeter, Exeter EX4 4PS, UK;
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, Hatherly Laboratories, University of Exeter, Exeter EX4 4PS, UK;
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
| |
Collapse
|
31
|
Vogel P, Ding ZM, Read R, DaCosta CM, Hansard M, Small DL, Ye GL, Hansen G, Brommage R, Powell DR. Progressive Degenerative Myopathy and Myosteatosis in ASNSD1-Deficient Mice. Vet Pathol 2020; 57:723-735. [PMID: 32638637 DOI: 10.1177/0300985820939251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mice with an inactivating mutation in the gene encoding asparagine synthetase domain containing 1 (ASNSD1) develop a progressive degenerative myopathy that results in severe sarcopenia and myosteatosis. ASNSD1 is conserved across many species, and whole body gene expression surveys show maximal expression levels of ASNSD1 in skeletal muscle. However, potential functions of this protein have not been previously reported. Asnsd1-/- mice demonstrated severe muscle weakness, and their normalized body fat percentage on both normal chow and high fat diets was greater than 2 SD above the mean for 3651 chow-fed and 2463 high-fat-diet-fed knockout (KO) lines tested. Histologic lesions were essentially limited to the muscle and were characterized by a progressive degenerative myopathy with extensive transdifferentiation and replacement of muscle by well-differentiated adipose tissue. There was minimal inflammation, fibrosis, and muscle regeneration associated with this myopathy. In addition, the absence of any signs of lipotoxicity in Asnsd1-/- mice despite their extremely elevated body fat percentage and low muscle mass suggests a role for metabolic dysfunctions in the development of this phenotype. Asnsd1-/- mice provide the first insight into the function of this protein, and this mouse model could prove useful in elucidating fundamental metabolic interactions between skeletal muscle and adipose tissue.
Collapse
Affiliation(s)
- Peter Vogel
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | - Zhi-Ming Ding
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | - Robert Read
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | | | | | - Daniel L Small
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | - Gui-Lan Ye
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | - Gwenn Hansen
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| | | | - David R Powell
- 57636Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW The apical Na/K/2Cl cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb, contributing to maintenance of blood pressure (BP). Despite effective NKCC2 inhibition by loop diuretics, these agents are not viable for long-term management of BP due to side effects. Novel molecular mechanisms that control NKCC2 activity reveal an increasingly complex picture with interacting layers of NKCC2 regulation. Here, we review the latest developments that shine new light on NKCC2-mediated control of BP and potential new long-term therapies to treat hypertension. RECENT FINDINGS Emerging molecular NKCC2 regulators, often binding partners, reveal a complex overlay of interacting mechanisms aimed at fine tuning NKCC2 activity. Different factors achieve this by shifting the balance between trafficking steps like exocytosis, endocytosis, recycling and protein turnover, or by balancing phosphorylation vs. dephosphorylation. Further molecular details are also emerging on previously known pathways of NKCC2 regulation, and recent in-vivo data continues to place NKCC2 regulation at the center of BP control. SUMMARY Several layers of emerging molecular mechanisms that control NKCC2 activity may operate simultaneously, but they can also be controlled independently. This provides an opportunity to identify new pharmacological targets to fine-tune NKCC2 activity for BP management.
Collapse
|
33
|
Prins KW, Kalra R, Rose L, Assad TR, Archer SL, Bajaj NS, Weir EK, Prisco SZ, Pritzker M, Lutsey PL, Brittain EL, Thenappan T. Hypochloremia Is a Noninvasive Predictor of Mortality in Pulmonary Arterial Hypertension. J Am Heart Assoc 2020; 9:e015221. [PMID: 32079477 PMCID: PMC7335577 DOI: 10.1161/jaha.119.015221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Pulmonary arterial hypertension (PAH) is a lethal disease. In resource‐limited countries PAH outcomes are worse because therapy costs are prohibitive. To improve global outcomes, noninvasive and widely available biomarkers that identify high‐risk patients should be defined. Serum chloride is widely available and predicts mortality in left heart failure, but its prognostic utility in PAH requires further investigation. Methods and Results In this study 475 consecutive PAH patients evaluated at the University of Minnesota and Vanderbilt University PAH clinics were examined. Clinical characteristics were compared by tertiles of serum chloride. Both the Kaplan‐Meier method and Cox regression analysis were used to assess survival and predictors of mortality, respectively. Categorical net reclassification improvement and relative integrated discrimination improvement compared prediction models. PAH patients in the lowest serum chloride tertile (≤101 mmol/L: hypochloremia) had the lowest 6‐minute walk distance and highest right atrial pressure despite exhibiting no differences in pulmonary vascular disease severity. The 1‐, 3‐, and 5‐year survival was reduced in hypochloremic patients when compared with the middle‐ and highest‐tertile patients (86%/64%/44%, 95%/78%/59%, and, 91%/79%/66%). After adjustment for age, sex, diuretic use, serum sodium, bicarbonate, and creatinine, the hypochloremic patients had increased mortality when compared with the middle‐tertile and highest‐tertile patients. The Minnesota noninvasive model (functional class, 6‐minute walk distance, and hypochloremia) was as effective as the French noninvasive model (functional class, 6‐minute walk distance, and elevated brain natriuretic peptide or N‐terminal pro–brain natriuretic peptide) for predicting mortality. Conclusions Hypochloremia (≤101 mmol/L) identifies high‐risk PAH patients independent of serum sodium, renal function, and diuretic use.
Collapse
Affiliation(s)
- Kurt W Prins
- Cardiovascular Division University of Minnesota Minneapolis MN
| | - Rajat Kalra
- Cardiovascular Division University of Minnesota Minneapolis MN
| | - Lauren Rose
- Cardiovascular Division University of Minnesota Minneapolis MN
| | | | | | | | - E Kenneth Weir
- Cardiovascular Division University of Minnesota Minneapolis MN
| | - Sasha Z Prisco
- Cardiovascular Division University of Minnesota Minneapolis MN
| | - Marc Pritzker
- Cardiovascular Division University of Minnesota Minneapolis MN
| | - Pamela L Lutsey
- School of Public Health University of Minnesota Minneapolis MN
| | - Evan L Brittain
- Vanderbilt University Medical Center and Vanderbilt Translational and Clinical Cardiovascular Research Center Nashville TN
| | | |
Collapse
|
34
|
Powell DR, Doree DD, DaCosta CM, Platt KA, Hansen GM, van Sligtenhorst I, Ding ZM, Revelli JP, Brommage R. Obesity of G2e3 Knockout Mice Suggests That Obesity-Associated Variants Near Human G2E3 Decrease G2E3 Activity. Diabetes Metab Syndr Obes 2020; 13:2641-2652. [PMID: 32801815 PMCID: PMC7394505 DOI: 10.2147/dmso.s259546] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE In humans, single nucleotide polymorphisms (SNPs) near the adjacent protein kinase D1 (PRKD1) and G2/M-phase-specific E3 ubiquitin protein ligase (G2E3) genes on chromosome 14 are associated with obesity. To date, no published evidence links inactivation of either gene to changes in body fat. These two genes are also adjacent on mouse chromosome 12. Because obesity genes are highly conserved between humans and mice, we analyzed body fat in adult G2e3 and Prkd1 knockout (KO) mice to determine whether inactivating either gene leads to obesity in mice and, by inference, probably in humans. METHODS The G2e3 and Prkd1 KO lines were generated by gene trapping and by homologous recombination methodologies, respectively. Body fat was measured by DEXA in adult mice fed chow from weaning and by QMR in a separate cohort of mice fed high-fat diet (HFD) from weaning. Glucose homeostasis was evaluated with oral glucose tolerance tests (OGTTs) performed on adult mice fed HFD from weaning. RESULTS Body fat was increased in multiple cohorts of G2e3 KO mice relative to their wild-type (WT) littermates. When data from all G2e3 KO (n=32) and WT (n=31) mice were compared, KO mice showed increases of 11% in body weight (P<0.01), 65% in body fat (P<0.001), 48% in % body fat (P<0.001), and an insignificant 3% decrease in lean body mass. G2e3 KO mice were also glucose intolerant during an OGTT (P<0.05). In contrast, Prkd1 KO and WT mice had comparable body fat levels and glucose tolerance. CONCLUSION Significant obesity and glucose intolerance were observed in G2e3, but not Prkd1, KO mice. The conservation of obesity genes between mice and humans strongly suggests that the obesity-associated SNPs located near the human G2E3 and PRKD1 genes are linked to variants that decrease the amount of functional human G2E3.
Collapse
Affiliation(s)
- David R Powell
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
- Correspondence: David R Powell Lexicon Pharmaceuticals Inc., 8800 Technology Forest Place, The Woodlands, TX77381, USATel +1 281 863 3060Fax +1 281 863 8115 Email
| | - Deon D Doree
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
| | | | | | - Gwenn M Hansen
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
| | | | - Zhi-Ming Ding
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, 77381, USA
| | | | | |
Collapse
|
35
|
Rautureau Y, Deschambault V, Higgins MÈ, Rivas D, Mecteau M, Geoffroy P, Miquel G, Uy K, Sanchez R, Lavoie V, Brand G, Nault A, Williams PM, Suarez ML, Merlet N, Lapointe L, Duquette N, Gillis MA, Samami S, Mayer G, Pouliot P, Raignault A, Maafi F, Brodeur MR, Levesque S, Guertin MC, Dubé MP, Thorin É, Rhainds D, Rhéaume É, Tardif JC. ADCY9 (Adenylate Cyclase Type 9) Inactivation Protects From Atherosclerosis Only in the Absence of CETP (Cholesteryl Ester Transfer Protein). Circulation 2019; 138:1677-1692. [PMID: 29674325 DOI: 10.1161/circulationaha.117.031134] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Pharmacogenomic studies have shown that ADCY9 genotype determines the effects of the CETP (cholesteryl ester transfer protein) inhibitor dalcetrapib on cardiovascular events and atherosclerosis imaging. The underlying mechanisms responsible for the interactions between ADCY9 and CETP activity have not yet been determined. METHODS Adcy9-inactivated ( Adcy9Gt/Gt) and wild-type (WT) mice, that were or not transgenic for the CETP gene (CETPtg Adcy9Gt/Gt and CETPtg Adcy9WT), were submitted to an atherogenic protocol (injection of an AAV8 [adeno-associated virus serotype 8] expressing a PCSK9 [proprotein convertase subtilisin/kexin type 9] gain-of-function variant and 0.75% cholesterol diet for 16 weeks). Atherosclerosis, vasorelaxation, telemetry, and adipose tissue magnetic resonance imaging were evaluated. RESULTS Adcy9Gt/Gt mice had a 65% reduction in aortic atherosclerosis compared to WT ( P<0.01). CD68 (cluster of differentiation 68)-positive macrophage accumulation and proliferation in plaques were reduced in Adcy9Gt/Gt mice compared to WT animals ( P<0.05 for both). Femoral artery endothelial-dependent vasorelaxation was improved in Adcy9Gt/Gt mice (versus WT, P<0.01). Selective pharmacological blockade showed that the nitric oxide, cyclooxygenase, and endothelial-dependent hyperpolarization pathways were all responsible for the improvement of vasodilatation in Adcy9Gt/Gt ( P<0.01 for all). Aortic endothelium from Adcy9Gt/Gt mice allowed significantly less adhesion of splenocytes compared to WT ( P<0.05). Adcy9Gt/Gt mice gained more weight than WT with the atherogenic diet; this was associated with an increase in whole body adipose tissue volume ( P<0.01 for both). Feed efficiency was increased in Adcy9Gt/Gt compared to WT mice ( P<0.01), which was accompanied by prolonged cardiac RR interval ( P<0.05) and improved nocturnal heart rate variability ( P=0.0572). Adcy9 inactivation-induced effects on atherosclerosis, endothelial function, weight gain, adipose tissue volume, and feed efficiency were lost in CETPtg Adcy9Gt/Gt mice ( P>0.05 versus CETPtg Adcy9WT). CONCLUSIONS Adcy9 inactivation protects against atherosclerosis, but only in the absence of CETP activity. This atheroprotection may be explained by decreased macrophage accumulation and proliferation in the arterial wall, and improved endothelial function and autonomic tone.
Collapse
Affiliation(s)
- Yohann Rautureau
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Vanessa Deschambault
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Marie-Ève Higgins
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Daniel Rivas
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Mélanie Mecteau
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Pascale Geoffroy
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Géraldine Miquel
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Kurunradeth Uy
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Rocio Sanchez
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Véronique Lavoie
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Geneviève Brand
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Audrey Nault
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Pierre-Marc Williams
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Maria Laura Suarez
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Nolwenn Merlet
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Line Lapointe
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Natacha Duquette
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Marc-Antoine Gillis
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Samaneh Samami
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Gaétan Mayer
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.).,Faculty of Pharmacy (G. Mayer), Université de Montréal, Canada
| | | | - Adeline Raignault
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Foued Maafi
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Mathieu R Brodeur
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Sylvie Levesque
- Montreal Health Innovations Coordinating Centre, Montreal, Canada (S.L., M-C.G.)
| | - Marie-Claude Guertin
- Montreal Health Innovations Coordinating Centre, Montreal, Canada (S.L., M-C.G.)
| | - Marie-Pierre Dubé
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada (M-P.D.)
| | - Éric Thorin
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.).,Departments of Surgery (E.T.), Université de Montréal, Canada
| | - David Rhainds
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.)
| | - Éric Rhéaume
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.).,Medicine (E.R., J-C-.T.) of the Faculty of Medicine, Université de Montréal, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Canada (Y.R., V.D., M-E.H., D.R., M.M., P.G., G.Miquel, K.U., R.S., V.L., G.B., A.N., P-M.W., M.L.S., N.M., L.L., N.D., M-A.G., S.S., G.Mayer, A.R., F.M., M.R.B., E.T., D.R., E.R., J-C.T.).,Medicine (E.R., J-C-.T.) of the Faculty of Medicine, Université de Montréal, Canada
| |
Collapse
|
36
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
37
|
Zeng XL, Sun L, Zheng HQ, Wang GL, Du YH, Lv XF, Ma MM, Guan YY. Smooth muscle-specific TMEM16A expression protects against angiotensin II-induced cerebrovascular remodeling via suppressing extracellular matrix deposition. J Mol Cell Cardiol 2019; 134:131-143. [PMID: 31301303 DOI: 10.1016/j.yjmcc.2019.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022]
Abstract
Cerebrovascular remodeling is the leading factor for stroke and characterized by increased extracellular matrix deposition, migration and proliferation of vascular smooth muscle cells, and inhibition of their apoptosis. TMEM16A is an important component of Ca2+-activated Cl- channels. Previously, we showed that downregulation of TMEM16A in the basilar artery was negatively correlated with cerebrovascular remodeling during hypertension. However, it is unclear whether TMEM16A participates in angiotensin II (Ang II)-induced vascular remodeling in mice that have TMEM16A gene modification. In this study, we generated a transgenic mouse that overexpresses TMEM16A specifically in vascular smooth muscle cells. We observed that vascular remodeling in the basilar artery during Ang II-induced hypertension was significantly suppressed upon vascular smooth muscle-specific overexpression of TMEM16A relative to control mice. Specifically, we observed a large reduction in the deposition of fibronectin and collagen I. The expression of matrix metalloproteinases (MMP-2, MMP-9, and MMP-14), and tissue inhibitors of metalloproteinases (TIMP-1 and TIMP-2) were upregulated in the basilar artery during Ang II-induced hypertension, but this was suppressed upon overexpression of TMEM16A in blood vessels. Furthermore, TMEM16A overexpression alleviated the overactivity of the canonical TGF-β1/Smad3, and non-canonical TGF-β1/ERK and JNK pathways in the basilar artery during Ang II-induced hypertension. These in vivo results were similar to the results derived in vitro with basilar artery smooth muscle cells stimulated by Ang II. Moreover, we observed that the inhibitory effect of TMEM16A on MMPs was mediated by decreasing the activation of WNK1, which is a Cl--sensitive serine/threonine kinase. In conclusion, this study demonstrates that TMEM16A protects against cerebrovascular remodeling during hypertension by suppressing extracellular matrix deposition. We also showed that TMEM16A exerts this effect by reducing the expression of MMPs via inhibiting WNK1, and decreasing the subsequent activities of TGF-β1/Smad3, ERK, and JNK. Accordingly, our results suggest that TMEM16A may serve as a novel therapeutic target for vascular remodeling.
Collapse
Affiliation(s)
- Xue-Lin Zeng
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Lu Sun
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hua-Qing Zheng
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guan-Lei Wang
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Yan-Hua Du
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Xiao-Fei Lv
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Ming-Ming Ma
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Yong-Yuan Guan
- Department of Pharmacology, and Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
38
|
Manosroi W, Williams GH. Genetics of Human Primary Hypertension: Focus on Hormonal Mechanisms. Endocr Rev 2019; 40:825-856. [PMID: 30590482 PMCID: PMC6936319 DOI: 10.1210/er.2018-00071] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Abstract
Increasingly, primary hypertension is being considered a syndrome and not a disease, with the individual causes (diseases) having a common sign-an elevated blood pressure. To determine these causes, genetic tools are increasingly employed. This review identified 62 proposed genes. However, only 21 of them met our inclusion criteria: (i) primary hypertension, (ii) two or more supporting cohorts from different publications or within a single publication or one supporting cohort with a confirmatory genetically modified animal study, and (iii) 600 or more subjects in the primary cohort; when including our exclusion criteria: (i) meta-analyses or reviews, (ii) secondary and monogenic hypertension, (iii) only hypertensive complications, (iv) genes related to blood pressure but not hypertension per se, (v) nonsupporting studies more common than supporting ones, and (vi) studies that did not perform a Bonferroni or similar multiassessment correction. These 21 genes were organized in a four-tiered structure: distant phenotype (hypertension); intermediate phenotype [salt-sensitive (18) or salt-resistant (0)]; subintermediate phenotypes under salt-sensitive hypertension [normal renin (4), low renin (8), and unclassified renin (6)]; and proximate phenotypes (specific genetically driven hypertensive subgroup). Many proximate hypertensive phenotypes had a substantial endocrine component. In conclusion, primary hypertension is a syndrome; many proposed genes are likely to be false positives; and deep phenotyping will be required to determine the utility of genetics in the treatment of hypertension. However, to date, the positive genes are associated with nearly 50% of primary hypertensives, suggesting that in the near term precise, mechanistically driven treatment and prevention strategies for the specific primary hypertension subgroups are feasible.
Collapse
Affiliation(s)
- Worapaka Manosroi
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Endocrinology and Metabolism, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Gordon H Williams
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
39
|
Brommage R, Powell DR, Vogel P. Predicting human disease mutations and identifying drug targets from mouse gene knockout phenotyping campaigns. Dis Model Mech 2019; 12:dmm038224. [PMID: 31064765 PMCID: PMC6550044 DOI: 10.1242/dmm.038224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Two large-scale mouse gene knockout phenotyping campaigns have provided extensive data on the functions of thousands of mammalian genes. The ongoing International Mouse Phenotyping Consortium (IMPC), with the goal of examining all ∼20,000 mouse genes, has examined 5115 genes since 2011, and phenotypic data from several analyses are available on the IMPC website (www.mousephenotype.org). Mutant mice having at least one human genetic disease-associated phenotype are available for 185 IMPC genes. Lexicon Pharmaceuticals' Genome5000™ campaign performed similar analyses between 2000 and the end of 2008 focusing on the druggable genome, including enzymes, receptors, transporters, channels and secreted proteins. Mutants (4654 genes, with 3762 viable adult homozygous lines) with therapeutically interesting phenotypes were studied extensively. Importantly, phenotypes for 29 Lexicon mouse gene knockouts were published prior to observations of similar phenotypes resulting from homologous mutations in human genetic disorders. Knockout mouse phenotypes for an additional 30 genes mimicked previously published human genetic disorders. Several of these models have helped develop effective treatments for human diseases. For example, studying Tph1 knockout mice (lacking peripheral serotonin) aided the development of telotristat ethyl, an approved treatment for carcinoid syndrome. Sglt1 (also known as Slc5a1) and Sglt2 (also known as Slc5a2) knockout mice were employed to develop sotagliflozin, a dual SGLT1/SGLT2 inhibitor having success in clinical trials for diabetes. Clinical trials evaluating inhibitors of AAK1 (neuropathic pain) and SGLT1 (diabetes) are underway. The research community can take advantage of these unbiased analyses of gene function in mice, including the minimally studied 'ignorome' genes.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - Peter Vogel
- St. Jude Children's Research Hospital, Pathology, MS 250, Room C5036A, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
40
|
Gallolu Kankanamalage S, Karra AS, Cobb MH. WNK pathways in cancer signaling networks. Cell Commun Signal 2018; 16:72. [PMID: 30390653 PMCID: PMC6215617 DOI: 10.1186/s12964-018-0287-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background The with no lysine [K] (WNK) pathway consists of the structurally unique WNK kinases, their downstream target kinases, oxidative stress responsive (OSR)1 and SPS/Ste20-related proline-alanine-rich kinase (SPAK), and a multitude of OSR1/SPAK substrates including cation chloride cotransporters. Main body While the best known functions of the WNK pathway is regulation of ion transport across cell membranes, WNK pathway components have been implicated in numerous human diseases. The goal of our review is to draw attention to how this pathway and its components exert influence on the progression of cancer, specifically by detailing WNK signaling intersections with major cell communication networks and processes. Conclusion Here we describe how WNKs and associated proteins interact with and influence PI3K-AKT, TGF-β, and NF-κB signaling, as well as its unanticipated role in the regulation of angiogenesis.
Collapse
Affiliation(s)
- Sachith Gallolu Kankanamalage
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390-9041, USA
| | - Aroon S Karra
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390-9041, USA
| | - Melanie H Cobb
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390-9041, USA.
| |
Collapse
|
41
|
Kim J, Lei Y, Guo J, Kim SE, Wlodarczyk BJ, Cabrera RM, Lin YL, Nilsson TK, Zhang T, Ren A, Wang L, Yuan Z, Zheng YF, Wang HY, Finnell RH. Formate rescues neural tube defects caused by mutations in Slc25a32. Proc Natl Acad Sci U S A 2018; 115:4690-4695. [PMID: 29666258 PMCID: PMC5939102 DOI: 10.1073/pnas.1800138115] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Periconceptional folic acid (FA) supplementation significantly reduces the prevalence of neural tube defects (NTDs). Unfortunately, some NTDs are FA resistant, and as such, NTDs remain a global public health concern. Previous studies have identified SLC25A32 as a mitochondrial folate transporter (MFT), which is capable of transferring tetrahydrofolate (THF) from cellular cytoplasm to the mitochondria in vitro. Herein, we show that gene trap inactivation of Slc25a32 (Mft) in mice induces NTDs that are folate (5-methyltetrahydrofolate, 5-mTHF) resistant yet are preventable by formate supplementation. Slc25a32gt/gt embryos die in utero with 100% penetrant cranial NTDs. 5-mTHF supplementation failed to promote normal neural tube closure (NTC) in mutant embryos, while formate supplementation enabled the majority (78%) of knockout embryos to complete NTC. A parallel genetic study in human subjects with NTDs identified biallelic loss of function SLC25A32 variants in a cranial NTD case. These data demonstrate that the loss of functional Slc25a32 results in cranial NTDs in mice and has also been observed in a human NTD patient.
Collapse
Affiliation(s)
- Jimi Kim
- Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78723
| | - Yunping Lei
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin , Austin, TX 78723
| | - Jin Guo
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, 100700 Beijing, China
| | - Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin , Austin, TX 78723
| | - Bogdan J Wlodarczyk
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin , Austin, TX 78723
| | - Robert M Cabrera
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin , Austin, TX 78723
| | - Ying Linda Lin
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin , Austin, TX 78723
| | - Torbjorn K Nilsson
- Department of Medical Biosciences, Clinical Chemistry, Umea University, SE-90185 Umea, Sweden
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, 100700 Beijing, China
| | - Aiguo Ren
- Institute of Reproductive and Child Health, Peking University, 100191 Beijing, China
| | - Linlin Wang
- Institute of Reproductive and Child Health, Peking University, 100191 Beijing, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, 117004 Shenyang, China
| | - Yu-Fang Zheng
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences of Fudan University, 20043 Shanghai, China
- Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Institute of Reproduction & Development and Children's Hospital of Fudan University, 200011 Shanghai, China
| | - Hong-Yan Wang
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences of Fudan University, 20043 Shanghai, China;
- Key Laboratory of Reproduction Regulation of National Population and Family Planning Commission, Institute of Reproduction & Development and Children's Hospital of Fudan University, 200011 Shanghai, China
| | - Richard H Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin , Austin, TX 78723;
- Collaborative Innovation Center for Genetics & Development, School of Life Sciences, Fudan University, 200438 Shanghai, China
| |
Collapse
|
42
|
Rafael C, Soukaseum C, Baudrie V, Frère P, Hadchouel J. Consequences of SPAK inactivation on Hyperkalemic Hypertension caused by WNK1 mutations: evidence for differential roles of WNK1 and WNK4. Sci Rep 2018; 8:3249. [PMID: 29459793 PMCID: PMC5818654 DOI: 10.1038/s41598-018-21405-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/10/2018] [Indexed: 12/16/2022] Open
Abstract
Mutations of the gene encoding WNK1 [With No lysine (K) kinase 1] or WNK4 cause Familial Hyperkalemic Hypertension (FHHt). Previous studies have shown that the activation of SPAK (Ste20-related Proline/Alanine-rich Kinase) plays a dominant role in the development of FHHt caused by WNK4 mutations. The implication of SPAK in FHHt caused by WNK1 mutation has never been investigated. To clarify this issue, we crossed WNK1+/FHHt mice with SPAK knock-in mice in which the T-loop Thr243 residue was mutated to alanine to prevent activation by WNK kinases. We show that WNK1+/FHHT:SPAK243A/243A mice display an intermediate phenotype, between that of control and SPAK243A/243A mice, with normal blood pressure but hypochloremic metabolic alkalosis. NCC abundance and phosphorylation levels also decrease below the wild-type level in the double-mutant mice but remain higher than in SPAK243A/243A mice. This is different from what was observed in WNK4-FHHt mice in which SPAK inactivation completely restored the phenotype and NCC expression to wild-type levels. Although these results confirm that FHHt caused by WNK1 mutations is dependent on the activation of SPAK, they suggest that WNK1 and WNK4 play different roles in the distal nephron.
Collapse
Affiliation(s)
- Chloé Rafael
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.,Universités Paris-Descartes et Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Sorbonne Université, Paris, France.,INSERM UMR_S1155, Tenon Hospital, Paris, France
| | - Christelle Soukaseum
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.,Universités Paris-Descartes et Paris-Diderot, Sorbonne Paris Cité, Paris, France.,INSERM UMR_S1176, CHU de Bicêtre, Le Kremlin-Bicêtre, France
| | - Véronique Baudrie
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.,Universités Paris-Descartes et Paris-Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - Perrine Frère
- Sorbonne Université, Paris, France.,INSERM UMR_S1155, Tenon Hospital, Paris, France
| | - Juliette Hadchouel
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France. .,Universités Paris-Descartes et Paris-Diderot, Sorbonne Paris Cité, Paris, France. .,Sorbonne Université, Paris, France. .,INSERM UMR_S1155, Tenon Hospital, Paris, France.
| |
Collapse
|
43
|
Maeda Y, Kudo S, Tsushima K, Sato E, Kubota C, Kayamori A, Bochimoto H, Koga D, Torii S, Gomi H, Watanabe T, Hosaka M. Impaired Processing of Prohormones in Secretogranin III-Null Mice Causes Maladaptation to an Inadequate Diet and Stress. Endocrinology 2018; 159:1213-1227. [PMID: 29281094 DOI: 10.1210/en.2017-00636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 12/15/2017] [Indexed: 11/19/2022]
Abstract
Secretogranin III (SgIII), a member of the granin family, binds both to another granin, chromogranin A (CgA), and to a cholesterol-rich membrane that is destined for secretory granules (SGs). The knockdown of SgIII in adrenocorticotropic hormone (ACTH)-producing AtT-20 cells largely impairs the regulated secretion of CgA and ACTH. To clarify the physiological roles of SgIII in vivo, we analyzed hormone secretion and SG biogenesis in newly established SgIII-knockout (KO) mice. Although the SgIII-KO mice were viable and fertile and exhibited no overt abnormalities under ordinary rearing conditions, a high-fat/high-sucrose diet caused pronounced obesity in the mice. Furthermore, in the SgIII-KO mice compared with wild-type (WT) mice, the stimulated secretion of active insulin decreased substantially, whereas the storage of proinsulin increased in the islets. The plasma ACTH was also less elevated in the SgIII-KO mice than in the WT mice after chronic restraint stress, whereas the storage level of the precursor proopiomelanocortin in the pituitary gland was somewhat increased. These findings suggest that the lack of SgIII causes maladaptation of endocrine cells to an inadequate diet and stress by impairing the proteolytic conversion of prohormones in SGs, whereas SG biogenesis and the basal secretion of peptide hormones under ordinary conditions are ensured by the compensatory upregulation of other residual granins or factors.
Collapse
Affiliation(s)
- Yoshinori Maeda
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Saki Kudo
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Ken Tsushima
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Eri Sato
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Chisato Kubota
- Biosignal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Aika Kayamori
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| | - Hiroki Bochimoto
- Health Care Administration Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Daisuke Koga
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Japan
| | - Seiji Torii
- Biosignal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hiroshi Gomi
- Department of Veterinary Anatomy, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Tsuyoshi Watanabe
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Japan
| | - Masahiro Hosaka
- Department of Biotechnology, Laboratory of Molecular Life Sciences, Akita Prefectural University, Akita, Japan
| |
Collapse
|
44
|
Sominsky L, Goularte JF, Andrews ZB, Spencer SJ. Acylated Ghrelin Supports the Ovarian Transcriptome and Follicles in the Mouse: Implications for Fertility. Front Endocrinol (Lausanne) 2018; 9:815. [PMID: 30697193 PMCID: PMC6340924 DOI: 10.3389/fendo.2018.00815] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022] Open
Abstract
Ghrelin, an orexigenic gut-derived peptide, is gaining increasing attention due to its multifaceted role in a number of physiological functions, including reproduction. Ghrelin exists in circulation primarily as des-acylated and acylated ghrelin. Des-acyl ghrelin, until recently considered to be an inactive form of ghrelin, is now known to have independent physiological functionality. However, the relative contribution of acyl and des-acyl ghrelin to reproductive development and function is currently unknown. Here we used ghrelin-O-acyltransferase (GOAT) knockout (KO) mice that have no measurable levels of endogenous acyl ghrelin and chronically high levels of des-acyl ghrelin, to characterize how the developmental and life-long absence of acyl ghrelin affects ovarian development and reproductive capacity. We combined the assessment of markers of reproductive maturity and the capacity to breed with measures of ovarian morphometry, as well as with ovarian RNA sequencing analysis. Our data show that while GOAT KO mice retain the capacity to breed in young adulthood, there is a diminished number of ovarian follicles (per mm3) in the juvenile and adult ovaries, due to a significant reduction in the number of small follicles, particularly the primordial follicles. We also show pronounced specific changes in the ovarian transcriptome in the juvenile GOAT KO ovary, indicative of a potential for premature ovarian development. Collectively, these findings indicate that an absence of acyl ghrelin does not prevent reproductive success but that appropriate levels of acyl and des-acyl ghrelin may be necessary for optimal ovarian maturation.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- *Correspondence: Luba Sominsky
| | - Jeferson F. Goularte
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Zane B. Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Sarah J. Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Zhang YJ, Zheng HQ, Chen BY, Sun L, Ma MM, Wang GL, Guan YY. WNK1 is required for proliferation induced by hypotonic challenge in rat vascular smooth muscle cells. Acta Pharmacol Sin 2018; 39:35-47. [PMID: 28770829 DOI: 10.1038/aps.2017.56] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/07/2017] [Indexed: 12/20/2022] Open
Abstract
Hypotonic challenge evoked vascular cell proliferation through activation of volume-regulated Cl- channel (VRCC), leading to a decrease in the intracellular Cl- concentration ([Cl-]i). We hypothesize that the decrease in [Cl-]i may activate one or several Cl--sensitive kinases, resulting in a subsequent signaling cascade. In this study we demonstrated that WNK1, a Cl--sensitive kinase, was involved in VRCC-induced proliferative signaling pathway in A10 vascular smooth muscle cells in vitro. A10 cells were exposed to a hypotonic challenge (225 mosmol·kg-1·H20), which caused significantly increase in WNK1 phosphorylation without altering WNK1 protein expression. WNK1 overexpression significantly increased hypotonic-induced A10 cell proliferation, whereas silencing of WNK1 caused an opposite action. WNK1 mutation did not affect hypotonic-induced WNK1 phosphorylation and cell proliferation. Silencing of WNK1 caused cell cycle arrest at G0/G1 phase and prevented transition from G1 to S phase, whereas the WNK1 overexpression accelerated cell cycle transition from G1 to S phase. Silencing of WNK1 significantly inhibited cyclin D1/cyclin E1 expression and increased p27kip/p21cip expression. WNK1 overexpression significantly increased cyclin D1/cyclin E1 expression and reduced p27KIP/p21CIP expression. In addition, WNK1 knockdown or overexpression significantly attenuated or increased the hypotonic-induced phosphorylation of Akt and PI3K respectively.In conclusion, the reduction in [Cl-]i caused by hypotonic challenge-induced VRCC opening evokes WNK1 phosphorylation in A10 VSMCs, which mediates cell cycle transition from G0/G1 to S phase and proliferation through the PI3K-Akt signaling pathway.
Collapse
|
46
|
Boyd-Shiwarski CR, Shiwarski DJ, Roy A, Namboodiri HN, Nkashama LJ, Xie J, McClain KL, Marciszyn A, Kleyman TR, Tan RJ, Stolz DB, Puthenveedu MA, Huang CL, Subramanya AR. Potassium-regulated distal tubule WNK bodies are kidney-specific WNK1 dependent. Mol Biol Cell 2017; 29:499-509. [PMID: 29237822 PMCID: PMC6014176 DOI: 10.1091/mbc.e17-08-0529] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/27/2017] [Accepted: 12/06/2017] [Indexed: 01/18/2023] Open
Abstract
WNK bodies are large punctate membraneless cytosolic signaling foci that sequester WNK serine–threonine kinases and form in renal distal tubular epithelial cells during shifts in total body potassium balance. The assembly of these structures requires KS-WNK1, a truncated isoform of the WNK1 gene that is exclusively expressed in the distal tubule. 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.
Collapse
Affiliation(s)
- Cary R Boyd-Shiwarski
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Daniel J Shiwarski
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Ankita Roy
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Hima N Namboodiri
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Lubika J Nkashama
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Jian Xie
- Department of Internal Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Kara L McClain
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Allison Marciszyn
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Thomas R Kleyman
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261.,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Roderick J Tan
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | | | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Arohan R Subramanya
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 .,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261.,VA Pittsburgh Healthcare System, Pittsburgh, PA 15240
| |
Collapse
|
47
|
Vibhuti, Khan R, Sharma A, Jain S, Mohanty S, Prasad K. Intra-arterial transplantation of human bone marrow mesenchymal stem cells (hBMMSCs) improves behavioral deficits and alters gene expression in rodent stroke model. J Neurochem 2017; 143:722-735. [PMID: 29049855 DOI: 10.1111/jnc.14241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022]
Abstract
Stroke is a multi-factorial polygenic disease and is a major cause of death and adult disability. Administration of bone marrow stem cells protects ischemic rat brain by facilitating recovery of neurological functions. But the molecular mechanism of stem cells action and their effect on gene expression is not well explored. In this study, we have transplanted 1 × 106 human bone marrow mesenchymal stem cells (hBMMSCs) in middle cerebral artery occluded (MCAo) adult male Wistar rats through intracarotid artery route at 24 h after surgery. Motor behavioral tests (rotarod and open field) were performed to assess the changes in motor functions at day 0 and day1, 4, 8 and 14. The expression of studied genes at mRNA and protein level was quantified by using Q-PCR and western blotting, respectively. Further, we have assessed the methylation pattern of promoter of these genes by using methylation-specific PCR. Data were analyzed statistically and correlated. A significant improvement in behavioral deficits was observed in stem cells treated group after 14th day post stroke. Significantly (p < 0.05) increased mRNA and protein levels of brain derived neurotrophic factor and ANP genes in hBMMSCs treated group along with decrease in methylation level at their promoter was observed. On the other hand, significantly decreased mRNA and protein level of TSP1 and WNK1 in hBMMSCs treated group was observed. In conclusion, hBMMSCs administration significantly improves the behavioral deficits by improving motor and locomotor coordination. The promoter of TSP1 and WNK1 genes was found to be hyper-methylated in hBMMSCs group resulting in their decreased expression while the promoter of ANP and brain derived neurotrophic factor was found to be hypo-methylated. This study might shed a light on how hBMMSCs affect the gene expression by modulating methylation status.
Collapse
Affiliation(s)
- Vibhuti
- Department of Neurology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rehan Khan
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Suman Jain
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kameshwar Prasad
- Department of Neurology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
48
|
Strassman A, Schnütgen F, Dai Q, Jones JC, Gomez AC, Pitstick L, Holton NE, Moskal R, Leslie ER, von Melchner H, Beier DR, Bjork BC. Generation of a multipurpose Prdm16 mouse allele by targeted gene trapping. Dis Model Mech 2017; 10:909-922. [PMID: 28424158 PMCID: PMC5536910 DOI: 10.1242/dmm.029561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/18/2017] [Indexed: 12/17/2022] Open
Abstract
Gene trap mutagenesis is a powerful tool to create loss-of-function mutations in mice and other model organisms. Modifications of traditional gene trap cassettes, including addition of conditional features in the form of Flip-excision (FlEx) arrays to enable directional gene trap cassette inversions by Cre and Flpe site-specific recombinases, greatly enhanced their experimental potential. By taking advantage of these conditional gene trap cassettes, we developed a generic strategy for generating conditional mutations and validated this strategy in mice carrying a multipurpose allele of the Prdm16 transcription factor gene. We demonstrate that the gene trap insertion creates a null mutation replicating the Pierre Robin sequence-type cleft palate phenotype of other Prdm16 mutant mice. Consecutive breeding to Flpe and Emx1IREScre deleter mice spatially restricted Prdm16 loss to regions of the forebrain expressing the homeobox gene Emx1, demonstrating the utility of the technology for the analysis of tissue-specific gene functions. Summary: Described is the first targeting of an invertible gene trap to generate a conditional Prdm16 mouse allele and its use to assess phenotypic consequences of Prdm16 loss during craniofacial and brain development.
Collapse
Affiliation(s)
- Alexander Strassman
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Frank Schnütgen
- Department for Molecular Hematology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Qi Dai
- Department of Molecular Biosciences, The Wenner-Gren Institute, The University of Stockholm, SE-106 91 Stockholm, Sweden
| | - Jennifer C Jones
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Angela C Gomez
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Lenore Pitstick
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Nathan E Holton
- Department of Orthodontics, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Russell Moskal
- Department of Anatomy, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Erin R Leslie
- Department of Anatomy, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Harald von Melchner
- Department for Molecular Hematology, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - David R Beier
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Bryan C Bjork
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
49
|
Hadchouel J, Ellison DH, Gamba G. Regulation of Renal Electrolyte Transport by WNK and SPAK-OSR1 Kinases. Annu Rev Physiol 2016; 78:367-89. [PMID: 26863326 DOI: 10.1146/annurev-physiol-021115-105431] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of four genes responsible for pseudohypoaldosteronism type II, or familial hyperkalemic hypertension, which features arterial hypertension with hyperkalemia and metabolic acidosis, unmasked a complex multiprotein system that regulates electrolyte transport in the distal nephron. Two of these genes encode the serine-threonine kinases WNK1 and WNK4. The other two genes [kelch-like 3 (KLHL3) and cullin 3 (CUL3)] form a RING-type E3-ubiquitin ligase complex that modulates WNK1 and WNK4 abundance. WNKs regulate the activity of the Na(+):Cl(-) cotransporter (NCC), the epithelial sodium channel (ENaC), the renal outer medullary potassium channel (ROMK), and other transport pathways. Interestingly, the modulation of NCC occurs via the phosphorylation by WNKs of other serine-threonine kinases known as SPAK-OSR1. In contrast, the process of regulating the channels is independent of SPAK-OSR1. We present a review of the remarkable advances in this area in the past 10 years.
Collapse
Affiliation(s)
- Juliette Hadchouel
- INSERM UMR970, Paris Cardiovascular Research Center, 75015 Paris, France.,Faculty of Medicine, Paris Descartes University, Sorbonne Paris Cité, 75006 Paris, France
| | - David H Ellison
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City 14080, Mexico;
| |
Collapse
|
50
|
Murthy M, Kurz T, O'Shaughnessy KM. WNK signalling pathways in blood pressure regulation. Cell Mol Life Sci 2016; 74:1261-1280. [PMID: 27815594 PMCID: PMC5346417 DOI: 10.1007/s00018-016-2402-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/17/2016] [Accepted: 10/27/2016] [Indexed: 01/11/2023]
Abstract
Hypertension (high blood pressure) is a major public health problem affecting more than a billion people worldwide with complications, including stroke, heart failure and kidney failure. The regulation of blood pressure is multifactorial reflecting genetic susceptibility, in utero environment and external factors such as obesity and salt intake. In keeping with Arthur Guyton's hypothesis, the kidney plays a key role in blood pressure control and data from clinical studies; physiology and genetics have shown that hypertension is driven a failure of the kidney to excrete excess salt at normal levels of blood pressure. There is a number of rare Mendelian blood pressure syndromes, which have shed light on the molecular mechanisms involved in dysregulated ion transport in the distal kidney. One in particular is Familial hyperkalemic hypertension (FHHt), an autosomal dominant monogenic form of hypertension characterised by high blood pressure, hyperkalemia, hyperchloremic metabolic acidosis, and hypercalciuria. The clinical signs of FHHt are treated by low doses of thiazide diuretic, and it mirrors Gitelman syndrome which features the inverse phenotype of hypotension, hypokalemic metabolic alkalosis, and hypocalciuria. Gitelman syndrome is caused by loss of function mutations in the thiazide-sensitive Na/Cl cotransporter (NCC); however, FHHt patients do not have mutations in the SCL12A3 locus encoding NCC. Instead, mutations have been identified in genes that have revealed a key signalling pathway that regulates NCC and several other key transporters and ion channels in the kidney that are critical for BP regulation. This is the WNK kinase signalling pathway that is the subject of this review.
Collapse
Affiliation(s)
- Meena Murthy
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, CB2 2QQ, UK
| | - Thimo Kurz
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow, G12 8QQ, Scotland, UK
| | - Kevin M O'Shaughnessy
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, CB2 2QQ, UK.
| |
Collapse
|