1
|
Jaykumar AB, Monu SR, Mendez M, Rhaleb NE, Ortiz PA. ALMS1 KO rat: a new model of metabolic syndrome with spontaneous hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614364. [PMID: 39386593 PMCID: PMC11463523 DOI: 10.1101/2024.09.22.614364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
ALMS1 is a protein initially associated with Alström syndrome. This is a rare human disorder characterized by metabolic dysfunction, hypertension, obesity and hyperinsulinemia. In addition, ALMS1 gene was linked to hypertension status in a multipoint linkage population analysis. However, the mechanisms by which ALMS1 contributes to the development of obesity, insulin resistance and other metabolic disturbances are unknown. To study the role of ALMS1 in blood pressure regulation and renal function we previously generated an ALMS1 knockout rat model, where we found these rats are hypertensive. In this study, we further characterized the ALMS1 knockout rat, and found that they exhibit most characteristics of metabolic syndrome including hypertension and higher body weight by 10-12 weeks of age. In contrast, obesity, hyperinsulinemia and vascular dysfunction manifested at around 14-16 weeks of age. Interestingly, ALMS1 KO rats developed hyperleptinemia prior to the development of obesity rapidly after weaning by 7 weeks of age, suggesting an early role for ALMS1 in the hormonal control of leptin. We also found that female ALMS1 KO rats develop severe metabolic syndrome with hypertension similar to their male counterparts, lacking any protection often associated with better cardiovascular outcomes. Therefore, ALMS1 is an essential gene for sex- and age-dependent metabolic function. The ALMS1 knockout rat provides an invaluable pre-clinical animal model that recapitulates most symptoms present in patients and allows the study of new drugs and mechanisms that cause metabolic syndrome.
Collapse
Affiliation(s)
- Ankita B. Jaykumar
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, USA
- Department of Physiology, Wayne State School of Medicine, Detroit, USA
| | - Sumit R. Monu
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, USA
- Department of Physiology, Wayne State School of Medicine, Detroit, USA
| | - Mariela Mendez
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, USA
- Department of Physiology, Wayne State School of Medicine, Detroit, USA
| | - Nour-Eddine Rhaleb
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, USA
- Department of Physiology, Wayne State School of Medicine, Detroit, USA
| | - Pablo A. Ortiz
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, USA
- Department of Physiology, Wayne State School of Medicine, Detroit, USA
| |
Collapse
|
2
|
Jaykumar AB, Binns D, Taylor CA, Anselmo A, Birnbaum SG, Huber KM, Cobb MH. WNKs regulate mouse behavior and alter central nervous system glucose uptake and insulin signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598125. [PMID: 38915673 PMCID: PMC11195145 DOI: 10.1101/2024.06.09.598125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Certain areas of the brain involved in episodic memory and behavior, such as the hippocampus, express high levels of insulin receptors and glucose transporter-4 (GLUT4) and are responsive to insulin. Insulin and neuronal glucose metabolism improve cognitive functions and regulate mood in humans. Insulin-dependent GLUT4 trafficking has been extensively studied in muscle and adipose tissue, but little work has demonstrated either how it is controlled in insulin-responsive brain regions or its mechanistic connection to cognitive functions. In this study, we demonstrate that inhibition of WNK (With-No-lysine (K)) kinases improves learning and memory in mice. Neuronal inhibition of WNK enhances in vivo hippocampal glucose uptake. Inhibition of WNK enhances insulin signaling output and insulin-dependent GLUT4 trafficking to the plasma membrane in mice primary neuronal cultures and hippocampal slices. Therefore, we propose that the extent of neuronal WNK kinase activity has an important influence on learning, memory and anxiety-related behaviors, in part, by modulation of neuronal insulin signaling.
Collapse
Affiliation(s)
- Ankita B. Jaykumar
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Derk Binns
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Clinton A. Taylor
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Anthony Anselmo
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Shari G. Birnbaum
- Departments of Peter O’Donnell Jr. Brain Institute and Psychiatry, UT Southwestern Medical Center, Dallas, USA
| | | | - Melanie H. Cobb
- Departments of Pharmacology, UT Southwestern Medical Center, Dallas, USA
| |
Collapse
|
3
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Patel L, Roy A, Alvior AMB, Yuan M, Baig S, Bunting KV, Hodson J, Gehmlich K, Lord JM, Geberhiwot T, Steeds RP. Phenoage and longitudinal changes on transthoracic echocardiography in Alström syndrome: a disease of accelerated ageing? GeroScience 2024; 46:1989-1999. [PMID: 37782438 PMCID: PMC10828353 DOI: 10.1007/s11357-023-00959-3] [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: 09/06/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023] Open
Abstract
Alström syndrome (AS) is an ultra-rare disorder characterised by early-onset multi-organ dysfunction, such as insulin resistance, obesity, dyslipidaemia, and renal and cardiovascular disease. The objective is to explore whether AS is a disease of accelerated ageing and whether changes over time on echocardiography could reflect accelerated cardiac ageing. Cross-sectional measurement of Phenoage and retrospective analysis of serial echocardiography were performed between March 2012 and November 2022. The setting is a single national tertiary service jointly run by health service and patient charity. Forty-five adult patients aged over 16 years were included, 64% were male and 67% of White ethnicity. The median Phenoage was 48 years (interquartile range [IQR]: 35-72) in the 34 patients for whom this was calculable, which was significantly higher than the median chronological age of 29 years (IQR: 22-39, p<0.001). Phenoage was higher than chronological age in 85% (N=29) of patients, with a median difference of +18 years (IQR: +4, +34). On echocardiography, significant decreases were observed over time in left ventricular (LV) size at end-diastole (average of 0.046 cm per year, p<0.001) and end-systole (1.1% per year, p=0.025), with significant increase in posterior wall thickness at end-diastole (0.009 cm per year, p=0.008). LV systolic function measured by global longitudinal strain reduced (0.34 percentage points per year, p=0.020) and E/e'lat increased (2.5% per year, p=0.019). Most AS patients display a higher Phenoage compared to chronological age. Cardiac changes in AS patients were also reflective of accelerated ageing, with a reduction in LV size and increased wall thickening. AS may be a paradigm disease for premature ageing.
Collapse
Affiliation(s)
- Leena Patel
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Ashwin Roy
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK.
| | - Amor Mia B Alvior
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Mengshi Yuan
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Shanat Baig
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Karina V Bunting
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - James Hodson
- Research Development and Innovation, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Janet M Lord
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Tarekegn Geberhiwot
- Department of Endocrinology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, UK
| | - Richard P Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| |
Collapse
|
5
|
Benjamin JI, Pollock DM. Current perspective on circadian function of the kidney. Am J Physiol Renal Physiol 2024; 326:F438-F459. [PMID: 38134232 PMCID: PMC11207578 DOI: 10.1152/ajprenal.00247.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023] Open
Abstract
Behavior and function of living systems are synchronized by the 24-h rotation of the Earth that guides physiology according to time of day. However, when behavior becomes misaligned from the light-dark cycle, such as in rotating shift work, jet lag, and even unusual eating patterns, adverse health consequences such as cardiovascular or cardiometabolic disease can arise. The discovery of cell-autonomous molecular clocks expanded interest in regulatory systems that control circadian physiology including within the kidney, where function varies along a 24-h cycle. Our understanding of the mechanisms for circadian control of physiology is in the early stages, and so the present review provides an overview of what is known and the many gaps in our current understanding. We include a particular focus on the impact of eating behaviors, especially meal timing. A better understanding of the mechanisms guiding circadian function of the kidney is expected to reveal new insights into causes and consequences of a wide range of disorders involving the kidney, including hypertension, obesity, and chronic kidney disease.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
6
|
Woerz F, Hoffmann F, Antony S, Bolz S, Jarboui MA, Junger K, Klose F, Stehle IF, Boldt K, Ueffing M, Beyer T. Interactome Analysis Reveals a Link of the Novel ALMS1-CEP70 Complex to Centrosomal Clusters. Mol Cell Proteomics 2024; 23:100701. [PMID: 38122899 PMCID: PMC10820798 DOI: 10.1016/j.mcpro.2023.100701] [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: 06/02/2023] [Revised: 11/08/2023] [Accepted: 12/17/2023] [Indexed: 12/23/2023] Open
Abstract
Alström syndrome (ALMS) is a very rare autosomal-recessive disorder, causing a broad range of clinical defects most notably retinal degeneration, type 2 diabetes, and truncal obesity. The ALMS1 gene encodes a complex and huge ∼0.5 MDa protein, which has hampered analysis in the past. The ALMS1 protein is localized to the centrioles and the basal body of cilia and is involved in signaling processes, for example, TGF-β signaling. However, the exact molecular function of ALMS1 at the basal body remains elusive and controversial. We recently demonstrated that protein complex analysis utilizing endogenously tagged cells provides an excellent tool to investigate protein interactions of ciliary proteins. Here, CRISPR/Cas9-mediated endogenously tagged ALMS1 cells were used for affinity-based protein complex analysis. Centrosomal and microtubule-associated proteins were identified, which are potential regulators of ALMS1 function, such as the centrosomal protein 70 kDa (CEP70). Candidate proteins were further investigated in ALMS1-deficient hTERT-RPE1 cells. Loss of ALMS1 led to shortened cilia with no change in structural protein localization, for example, acetylated and ɣ-tubulin, Centrin-3, or the novel interactor CEP70. Conversely, reduction of CEP70 resulted in decreased ALMS1 at the ciliary basal body. Complex analysis of CEP70 revealed domain-specific ALMS1 interaction involving the TPR-containing C-terminal (TRP-CT) fragment of CEP70. In addition to ALMS1, several ciliary proteins, including CEP135, were found to specifically bind to the TPR-CT domain. Data are available via ProteomeXchange with the identifier PXD046401. Protein interactors identified in this study provide candidate lists that help to understand ALMS1 and CEP70 function in cilia-related protein modification, cell death, and disease-related mechanisms.
Collapse
Affiliation(s)
- Franziska Woerz
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.
| | - Felix Hoffmann
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Shibu Antony
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Sylvia Bolz
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Mohamed Ali Jarboui
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Katrin Junger
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Franziska Klose
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Isabel F Stehle
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Marius Ueffing
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Tina Beyer
- Eberhard Karls University Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
7
|
Bea-Mascato B, Gómez-Castañeda E, Sánchez-Corrales YE, Castellano S, Valverde D. Loss of the centrosomal protein ALMS1 alters lipid metabolism and the regulation of extracellular matrix-related processes. Biol Direct 2023; 18:84. [PMID: 38062477 PMCID: PMC10704752 DOI: 10.1186/s13062-023-00441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Alström syndrome (ALMS) is a rare autosomal recessive disease that is associated with mutations in ALMS1 gene. The main clinical manifestations of ALMS are retinal dystrophy, obesity, type 2 diabetes mellitus, dilated cardiomyopathy and multi-organ fibrosis, characteristic in kidneys and liver. Depletion of the protein encoded by ALMS1 has been associated with the alteration of different processes regulated via the primary cilium, such as the NOTCH or TGF-β signalling pathways. However, the cellular impact of these deregulated pathways in the absence of ALMS1 remains unknown. METHODS In this study, we integrated RNA-seq and proteomic analysis to determine the gene expression profile of hTERT-BJ-5ta ALMS1 knockout fibroblasts after TGF-β stimulation. In addition, we studied alterations in cross-signalling between the TGF-β pathway and the AKT pathway in this cell line. RESULTS We found that ALMS1 depletion affects the TGF-β pathway and its cross-signalling with other pathways such as PI3K/AKT, EGFR1 or p53. In addition, alterations associated with ALMS1 depletion clustered around the processes of extracellular matrix regulation and lipid metabolism in both the transcriptome and proteome. By studying the enriched pathways of common genes differentially expressed in the transcriptome and proteome, collagen fibril organisation, β-oxidation of fatty acids and eicosanoid metabolism emerged as key processes altered by the absence of ALMS1. Finally, an overactivation of the AKT pathway was determined in the absence of ALMS1 that could be explained by a decrease in PTEN gene expression. CONCLUSION ALMS1 deficiency disrupts cross-signalling between the TGF-β pathway and other dependent pathways in hTERT-BJ-5ta cells. Furthermore, altered cross-signalling impacts the regulation of extracellular matrix-related processes and fatty acid metabolism, and leads to over-activation of the AKT pathway.
Collapse
Affiliation(s)
- Brais Bea-Mascato
- CINBIO Facultad de Biología, Universidad de Vigo, Campus As Lagoas-Marcosende s/n, Vigo, 36310, Spain
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Eduardo Gómez-Castañeda
- Molecular and Cellular Immunology Section, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Yara E Sánchez-Corrales
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sergi Castellano
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK
- Zayed Centre for Research into Rare Disease in Children, UCL Genomics, University College London, London, UK
| | - Diana Valverde
- CINBIO Facultad de Biología, Universidad de Vigo, Campus As Lagoas-Marcosende s/n, Vigo, 36310, Spain.
- Grupo de Investigación en Enfermedades Raras y Medicina Pediátrica, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
8
|
Monu SR, Potter DL, Liao TD, King KN, Ortiz PA. Role of Alström syndrome 1 in the regulation of glomerular hemodynamics. Am J Physiol Renal Physiol 2023; 325:F418-F425. [PMID: 37560774 PMCID: PMC10639022 DOI: 10.1152/ajprenal.00017.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
Inactivating mutations in the ALMS1 gene in humans cause Alström syndrome, characterized by the early onset of obesity, insulin resistance, and renal dysfunction. However, the role of ALMS1 in renal function and hemodynamics is unclear. We previously found that ALMS1 is expressed in thick ascending limbs, where it binds and decreases Na+-K+-2Cl- cotransporter activity. We hypothesized that ALMS1 is expressed in macula densa cells and that its deletion enhances tubuloglomerular feedback (TGF) and reduces glomerular filtration rate (GFR) in rats. To test this, homozygous ALMS1 knockout (KO) and littermate wild-type Dahl salt-sensitive rats were studied. TGF sensitivity was higher in ALMS1 KO rats as measured by in vivo renal micropuncture. Using confocal microscopy, we confirmed immunolabeling of ALMS1 in macula densa cells (nitric oxide synthase 1 positive), supporting a role for ALMS1 in TGF regulation. Baseline glomerular capillary pressure was higher in ALMS1 KO rats, as was mean arterial pressure. Renal interstitial hydrostatic pressure was lower in ALMS1 KO rats, which is linked to increased Na+ reabsorption and hypertension. GFR was reduced in ALMS1 KO rats. Seven-week-old ALMS1 KO rats were not proteinuric, but proteinuria was present in 18- to 22-wk-old ALMS1 KO rats. The glomerulosclerosis index was higher in 18-wk-old ALMS1 KO rats. In conclusion, ALMS1 is involved in the control of glomerular hemodynamics in part by enhancing TGF sensitivity, and this may contribute to decreased GFR. Increased TGF sensitivity, enhanced glomerular capillary pressure, and hypertension may lead to glomerular damage in ALMS1 KO rats. These are the first data supporting the role of ALMS1 in TGF and glomerular hemodynamics.NEW & NOTEWORTHY ALMS1 is a novel protein involved in regulating tubuloglomerular feedback (TGF) sensitivity, glomerular capillary pressure, and blood pressure, and its dysfunction may reduce renal function and cause glomerular damage.
Collapse
Affiliation(s)
- Sumit R Monu
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, United States
- Department of Physiology, Wayne State University, Detroit, Michigan, United States
| | - D'Anna L Potter
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, United States
| | - Tang-Dong Liao
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, United States
| | - Keyona Nicole King
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, United States
| | - Pablo A Ortiz
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, United States
- Department of Physiology, Wayne State University, Detroit, Michigan, United States
| |
Collapse
|
9
|
Reynolds KM, Horimoto ARVR, Lin BM, Zhang Y, Kurniansyah N, Yu B, Boerwinkle E, Qi Q, Kaplan R, Daviglus M, Hou L, Zhou LY, Cai J, Shaikh SR, Sofer T, Browning SR, Franceschini N. Ancestry-driven metabolite variation provides insights into disease states in admixed populations. Genome Med 2023; 15:52. [PMID: 37461045 PMCID: PMC10351197 DOI: 10.1186/s13073-023-01209-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: 11/22/2022] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Metabolic pathways are related to physiological functions and disease states and are influenced by genetic variation and environmental factors. Hispanics/Latino individuals have ancestry-derived genomic regions (local ancestry) from their recent admixture that have been less characterized for associations with metabolite abundance and disease risk. METHODS We performed admixture mapping of 640 circulating metabolites in 3887 Hispanic/Latino individuals from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Metabolites were quantified in fasting serum through non-targeted mass spectrometry (MS) analysis using ultra-performance liquid chromatography-MS/MS. Replication was performed in 1856 nonoverlapping HCHS/SOL participants with metabolomic data. RESULTS By leveraging local ancestry, this study identified significant ancestry-enriched associations for 78 circulating metabolites at 484 independent regions, including 116 novel metabolite-genomic region associations that replicated in an independent sample. Among the main findings, we identified Native American enriched genomic regions at chromosomes 11 and 15, mapping to FADS1/FADS2 and LIPC, respectively, associated with reduced long-chain polyunsaturated fatty acid metabolites implicated in metabolic and inflammatory pathways. An African-derived genomic region at chromosome 2 was associated with N-acetylated amino acid metabolites. This region, mapped to ALMS1, is associated with chronic kidney disease, a disease that disproportionately burdens individuals of African descent. CONCLUSIONS Our findings provide important insights into differences in metabolite quantities related to ancestry in admixed populations including metabolites related to regulation of lipid polyunsaturated fatty acids and N-acetylated amino acids, which may have implications for common diseases in populations.
Collapse
Affiliation(s)
- Kaylia M Reynolds
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina, 123 W Franklin St, Suite 401, NC, NC 27516, Chapel Hill, USA
| | | | - Bridget M Lin
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Ying Zhang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Laura Y Zhou
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Saame Raza Shaikh
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Departments of Medicine and Biostatistics, Harvard University, Boston, MA, USA
| | - Sharon R Browning
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, 123 W Franklin St, Suite 401, NC, NC 27516, Chapel Hill, USA.
| |
Collapse
|
10
|
Costello HM, Johnston JG, Juffre A, Crislip GR, Gumz ML. Circadian clocks of the kidney: function, mechanism, and regulation. Physiol Rev 2022; 102:1669-1701. [PMID: 35575250 PMCID: PMC9273266 DOI: 10.1152/physrev.00045.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022] Open
Abstract
An intrinsic cellular circadian clock is located in nearly every cell of the body. The peripheral circadian clocks within the cells of the kidney contribute to the regulation of a variety of renal processes. In this review, we summarize what is currently known regarding the function, mechanism, and regulation of kidney clocks. Additionally, the effect of extrarenal physiological processes, such as endocrine and neuronal signals, on kidney function is also reviewed. Circadian rhythms in renal function are an integral part of kidney physiology, underscoring the importance of considering time of day as a key biological variable. The field of circadian renal physiology is of tremendous relevance, but with limited physiological and mechanistic information on the kidney clocks this is an area in need of extensive investigation.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|
11
|
Abstract
The BBSome is an octameric protein complex involved in Bardet-Biedl syndrome (BBS), a human pleiotropic, autosomal recessive condition. Patients with BBS display various clinical features including obesity, hypertension, and renal abnormalities. Association studies have also linked the BBS genes to hypertension and other cardiovascular risks in the general population. The BBSome was originally associated with the function of cilia, a highly specialized organelle that extend from the cell membrane of most vertebrate cells. However, subsequent studies have implicated the BBSome in the control of a myriad of other cellular processes not related to cilia including cell membrane localization of receptors and gene expression. The development of animal models of BBS such as mouse lines lacking various components of the BBSome and associated proteins has facilitated studying their role in the control of cardiovascular function and deciphering the pathophysiological mechanisms responsible for the cardiovascular aberrations associated with BBS. These studies revealed the importance of the neuronal, renal, vascular, and cardiac BBSome in the regulation of blood pressure, renal function, vascular reactivity, and cardiac development. The BBSome has also emerged as a critical regulator of key systems involved in cardiovascular control including the renin-angiotensin system. Better understanding of the influence of the BBSome on the molecular and physiological processes relevant to cardiovascular health and disease has the potential of identifying novel mechanisms underlying hypertension and other cardiovascular risks.
Collapse
Affiliation(s)
- Yuying Zhao
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Human Toxicology Graduate Program, University of Iowa Graduate College, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Iowa City VA Health Care System, Iowa City, IA, USA,Corresponding author: Kamal Rahmouni, Ph.D., Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA, , Tel: 319 353 5256, Fax: 319 353 5350
| |
Collapse
|
12
|
Karlsson Linnér R, Mallard TT, Barr PB, Sanchez-Roige S, Madole JW, Driver MN, Poore HE, de Vlaming R, Grotzinger AD, Tielbeek JJ, Johnson EC, Liu M, Rosenthal SB, Ideker T, Zhou H, Kember RL, Pasman JA, Verweij KJH, Liu DJ, Vrieze S, Kranzler HR, Gelernter J, Harris KM, Tucker-Drob EM, Waldman ID, Palmer AA, Harden KP, Koellinger PD, Dick DM. Multivariate analysis of 1.5 million people identifies genetic associations with traits related to self-regulation and addiction. Nat Neurosci 2021; 24:1367-1376. [PMID: 34446935 PMCID: PMC8484054 DOI: 10.1038/s41593-021-00908-3] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Behaviors and disorders related to self-regulation, such as substance use, antisocial behavior and attention-deficit/hyperactivity disorder, are collectively referred to as externalizing and have shared genetic liability. We applied a multivariate approach that leverages genetic correlations among externalizing traits for genome-wide association analyses. By pooling data from ~1.5 million people, our approach is statistically more powerful than single-trait analyses and identifies more than 500 genetic loci. The loci were enriched for genes expressed in the brain and related to nervous system development. A polygenic score constructed from our results predicts a range of behavioral and medical outcomes that were not part of genome-wide analyses, including traits that until now lacked well-performing polygenic scores, such as opioid use disorder, suicide, HIV infections, criminal convictions and unemployment. Our findings are consistent with the idea that persistent difficulties in self-regulation can be conceptualized as a neurodevelopmental trait with complex and far-reaching social and health correlates.
Collapse
Affiliation(s)
| | - Travis T Mallard
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Peter B Barr
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James W Madole
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Morgan N Driver
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Holly E Poore
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Ronald de Vlaming
- Department of Economics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Jorim J Tielbeek
- Department of Complex Trait Genetics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mengzhen Liu
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, VA CT Healthcare System, West Haven, CT, USA
| | - Rachel L Kember
- Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Mental Illness Research Education and Clinical Center, Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Joëlle A Pasman
- Behavioural Science Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Karin J H Verweij
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dajiang J Liu
- Department of Public Health Sciences, Penn State University, Hershey, PA, USA
- Institute of Personalized Medicine, Penn State University, Hershey, PA, USA
| | - Scott Vrieze
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Henry R Kranzler
- Center for Studies of Addiction, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Mental Illness Research Education and Clinical Center, Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, VA CT Healthcare System, West Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, West Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, West Haven, CT, USA
| | - Kathleen Mullan Harris
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elliot M Tucker-Drob
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| | - Irwin D Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
- Center for Computational and Quantitative Genetics, Emory University, Atlanta, GA, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - K Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, University of Texas at Austin, Austin, TX, USA
| | - Philipp D Koellinger
- Department of Economics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA.
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
13
|
Guo F, Seldin M, Péterfy M, Charugundla S, Zhou Z, Lee SD, Mouton A, Rajbhandari P, Zhang W, Pellegrini M, Tontonoz P, Lusis AJ, Shih DM. NOTUM promotes thermogenic capacity and protects against diet-induced obesity in male mice. Sci Rep 2021; 11:16409. [PMID: 34385484 PMCID: PMC8361163 DOI: 10.1038/s41598-021-95720-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
We recently showed that NOTUM, a liver-secreted Wnt inhibitor, can acutely promote browning of white adipose. We now report studies of chronic overexpression of NOTUM in liver indicating that it protects against diet-induced obesity and improves glucose homeostasis in mice. Adeno-associated virus (AAV) vectors were used to overexpress GFP or mouse Notum in the livers of male C57BL/6J mice and the mice were fed an obesifying diet. After 14 weeks of high fat, high sucrose diet feeding, the AAV-Notum mice exhibited decreased obesity and improved glucose tolerance compared to the AAV-GFP mice. Gene expression and immunoblotting analysis of the inguinal fat and brown fat revealed increased expression of beige/brown adipocyte markers in the AAV-Notum group, suggesting enhanced thermogenic capacity by NOTUM. A β3 adrenergic receptor agonist-stimulated lipolysis test suggested increased lipolysis capacity by NOTUM. The levels of collagen and C–C motif chemokine ligand 2 (CCL2) in the epididymal white adipose tissue of the AAV-Notum mice were significantly reduced, suggesting decreased fibrosis and inflammation, respectively. RNA sequencing analysis of inguinal white adipose of 4-week chow diet-fed mice revealed a highly significant enrichment of extracellular matrix (ECM) functional cluster among the down-regulated genes in the AAV-Notum group, suggesting a potential mechanism contributing to improved glucose homeostasis. Our in vitro studies demonstrated that recombinant human NOTUM protein blocked the inhibitory effects of WNT3A on brown adipocyte differentiation. Furthermore, NOTUM attenuated WNT3A’s effects on upregulation of TGF-β signaling and its downstream targets. Overall, our data suggest that NOTUM modulates adipose tissue function by promoting thermogenic capacity and inhibiting fibrosis through inhibition of Wnt signaling.
Collapse
Affiliation(s)
- Fangfei Guo
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Marcus Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Miklós Péterfy
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Sarada Charugundla
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Zhiqiang Zhou
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Stephen D Lee
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alice Mouton
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, 90095, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine Mount Sinai, New York, NY, 10029, USA
| | - Wenchao Zhang
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Department of Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Matteo Pellegrini
- Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Aldons J Lusis
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Diana M Shih
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA.
| |
Collapse
|
14
|
Gatticchi L, Miertus J, Maltese PE, Bressan S, De Antoni L, Podracká L, Piteková L, Rísová V, Mällo M, Jaakson K, Joost K, Colombo L, Bertelli M. A very early diagnosis of Alstrӧm syndrome by next generation sequencing. BMC MEDICAL GENETICS 2020; 21:173. [PMID: 32867697 PMCID: PMC7460749 DOI: 10.1186/s12881-020-01110-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022]
Abstract
Background Alström syndrome is a rare recessively inherited disorder caused by variants in the ALMS1 gene. It is characterized by multiple organ dysfunction, including cone-rod retinal dystrophy, dilated cardiomyopathy, hearing loss, obesity, insulin resistance, hyperinsulinemia, type 2 diabetes mellitus and systemic fibrosis. Heterogeneity and age-dependent development of clinical manifestations make it difficult to obtain a clear diagnosis, especially in pediatric patients. Case presentation Here we report the case of a girl with Alström syndrome. Genetic examination was proposed at age 22 months when suspected macular degeneration was the only major finding. Next generation sequencing of a panel of genes linked to eye-related pathologies revealed two compound heterozygous variants in the ALMS1 gene. Frameshift variants c.1196_1202del, p.(Thr399Lysfs*11), rs761292021 and c.11310_11313del, (p.Glu3771Trpfs*18), rs747272625 were detected in exons 5 and 16, respectively. Both variants cause frameshifts and generation of a premature stop-codon that probably leads to mRNA nonsense-mediated decay. Validation and segregation of ALMS1 variants were confirmed by Sanger sequencing. Conclusions Genetic testing makes it possible, even in childhood, to increase the number of correct diagnoses of patients who have ambiguous phenotypes caused by rare genetic variants. The development of high-throughput sequencing technologies offers an exceptionally valuable screening tool for clear genetic diagnoses and ensures early multidisciplinary management and treatment of the emerging symptoms.
Collapse
Affiliation(s)
- Leonardo Gatticchi
- Department of Experimental Medicine, Laboratory of Biochemistry, University of Perugia, Perugia, Italy
| | - Jan Miertus
- Génius n. o, Trnava, Slovakia.,MAGI's Lab, Genetic Testing Laboratory, Via Delle Maioliche 57/D, 38068, Rovereto, TN, Italy
| | - Paolo Enrico Maltese
- MAGI's Lab, Genetic Testing Laboratory, Via Delle Maioliche 57/D, 38068, Rovereto, TN, Italy.
| | - Simone Bressan
- MAGI's Lab, Genetic Testing Laboratory, Via Delle Maioliche 57/D, 38068, Rovereto, TN, Italy
| | - Luca De Antoni
- MAGI Euregio, Via Maso della Pieve, 60/A, 39100, Bolzano, Italy
| | - Ludmila Podracká
- Department of Pediatrics, National Institute for Sick Children, Commenius University, Bratislava, Slovakia
| | - Lucia Piteková
- Department of Pediatrics, National Institute for Sick Children, Commenius University, Bratislava, Slovakia
| | - Vanda Rísová
- Institute of Histology and Embryology, Faculty of Medicine, Commenius University, Bratislava, Slovakia
| | | | | | | | - Leonardo Colombo
- Department of Ophthalmology, San Paolo Hospital, University of Milan, Milan, Italy
| | - Matteo Bertelli
- MAGI's Lab, Genetic Testing Laboratory, Via Delle Maioliche 57/D, 38068, Rovereto, TN, Italy.,MAGI Euregio, Via Maso della Pieve, 60/A, 39100, Bolzano, Italy
| |
Collapse
|
15
|
Kim YC, Ganguly S, Nespoux J, Freeman B, Zhang H, Brenner D, Dhar D, Vallon V. Western Diet Promotes Renal Injury, Inflammation, and Fibrosis in a Murine Model of Alström Syndrome. Nephron Clin Pract 2020; 144:400-412. [PMID: 32629454 DOI: 10.1159/000508636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/12/2020] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Alström syndrome is a rare recessive genetic disease caused by mutations in ALMS1, which encodes a protein that is related to cilia function and intracellular endosome trafficking. The syndrome has been linked to impaired glucose metabolism and CKD. Polymorphisms in Alms1 have likewise been linked to CKD, but little is known about the modification of the phenotype by environmental factors. METHODS To gain further insights, the fat aussie (foz) mouse strain, a genetic murine model of Alström syndrome, was exposed to a normal chow (NC) or to a Western diet (WD, 20% fat, 34% sucrose by weight, and 0.2% cholesterol) and renal outcomes were measured. RESULTS Body weight and albuminuria were higher in foz than in wild-type (WT) mice on both diets but WD significantly increased the difference. Measurement of plasma creatinine and cystatin C indicated that glomerular filtration rate was preserved in foz versus WT independent of diet. Renal markers of injury, inflammation, and fibrosis were similar in both genotypes on NC but significantly greater in foz than in WT mice on WD. A glucose tolerance test performed in foz and WT mice on WD revealed similar basal blood glucose levels and subsequent blood glucose profiles. CONCLUSIONS WD sensitizes a murine model of Alström syndrome to kidney injury, inflammation, and fibrosis, an effect that may not be solely due to effects on glucose metabolism. Polymorphisms in Alms1 may induce CKD in part by modulating the deleterious effects of high dietary fat and sucrose on kidney outcome.
Collapse
Affiliation(s)
- Young Chul Kim
- Division of Nephrology & Hypertension, Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA
| | - Souradipta Ganguly
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Josselin Nespoux
- Division of Nephrology & Hypertension, Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA
| | - Brent Freeman
- Division of Nephrology & Hypertension, Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA
| | - Haiyan Zhang
- Division of Nephrology & Hypertension, Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA
| | - David Brenner
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Debanjan Dhar
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Volker Vallon
- Division of Nephrology & Hypertension, Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA,
| |
Collapse
|
16
|
Zhu X, Li G, Li S, Gong Z, Liu J, Song S. Neutrophil-to-lymphocyte ratio and red blood cell distribution width-to-platelet ratio predict cardiovascular events in hemodialysis patients. Exp Ther Med 2020; 20:1105-1114. [PMID: 32742350 DOI: 10.3892/etm.2020.8756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases are among the primary causes of decreased quality of life as well as mortality of hemodialysis patients with end-stage renal disease. The aim of the present study was to evaluate the predictive value of the red blood cell distribution width (RDW)-to-platelet ratio (RPR) and neutrophil-to-lymphocyte ratio (NLR) regarding the occurrence or development of cardiovascular events in hemodialysis patients, as well as the prognostic value of this metric. A total of 219 hemodialysis patients with cardiovascular events (HCE group) and 276 hemodialysis patients with no cardiovascular events (HNCE group) were enrolled in the present study. The clinical characteristics and laboratory parameters on admission, including RDW, as well as neutrophil, lymphocyte and platelet counts, were recorded. The NLR and RPR were increased in the HCE group compared with those in the HNCE group and there was a positive association between the NLR or RPR and the incidence of cardiovascular events in hemodialysis patients. In the receiver operating characteristics curve analysis, the area under the curve of the RPR for predicting cardiovascular events in hemodialysis patients was 0.88, while that for the NLR was 0.84. The sensitivity and specificity of the RPR for predicting cardiovascular events in hemodialysis patients were 0.87 and 0.82 respectively, and for the NLR, they were 0.75 and 0.79, respectively. The RPR was an independent risk factor for the prognosis regarding cardiovascular events in hemodialysis patients. In addition, the NLR and RPR were correlated with brain natriuretic peptide (BNP), cardiac troponin I (cTnI), creatine kinase isoenzyme-MB (CK-MB), and associated with ST segment changes in HCE patients. In conclusion, it was possible to predict the incidence of cardiovascular events in hemodialysis patients using the NLR and RPR, while the RPR had a better sensitivity and specificity than the NLR. The RPR was an independent risk factor for the prognosis regarding cardiovascular events in hemodialysis patients. These routinely available parameters should be considered as novel diagnostic markers for the occurrence and development of cardiovascular events in hemodialysis patients and their prognosis.
Collapse
Affiliation(s)
- Xiangjun Zhu
- Department of Nephrology, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224005, P.R. China
| | - Gongqi Li
- Department of Clinical Laboratory, Linyi Traditional Hospital, Linyi, Shandong 276003, P.R. China
| | - Shujuan Li
- Department of Nephrology, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224005, P.R. China
| | - Zhuang Gong
- Department of Nephrology, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224005, P.R. China
| | - Jun Liu
- Department of Laboratory Medicine, The Fifth People's Hospital of Wuxi Affiliated to Jiangnan University, Wuxi, Jiangsu 214005, P.R. China
| | - Shu Song
- Department of Pathology, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224005, P.R. China
| |
Collapse
|
17
|
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
|
18
|
King-Medina KN, Romero CA. New molecular motif contributes to NKCC2 trafficking. J Physiol 2019; 597:5331-5332. [PMID: 31541451 DOI: 10.1113/jp278896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Keyona N King-Medina
- Department of Internal Medicine Renal Division, Emory University School of Medicine, Atlanta, GA, USA
| | - Cesar A Romero
- Department of Internal Medicine Renal Division, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
19
|
Grochowsky A, Gunay-Aygun M. Clinical characteristics of individual organ system disease in non-motile ciliopathies. TRANSLATIONAL SCIENCE OF RARE DISEASES 2019; 4:1-23. [PMID: 31763176 PMCID: PMC6864414 DOI: 10.3233/trd-190033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Non-motile ciliopathies (disorders of the primary cilia) include autosomal dominant and recessive polycystic kidney diseases, nephronophthisis, as well as multisystem disorders Joubert, Bardet-Biedl, Alström, Meckel-Gruber, oral-facial-digital syndromes, and Jeune chondrodysplasia and other skeletal ciliopathies. Chronic progressive disease of the kidneys, liver, and retina are common features in non-motile ciliopathies. Some ciliopathies also manifest neurological, skeletal, olfactory and auditory defects. Obesity and type 2 diabetes mellitus are characteristic features of Bardet-Biedl and Alström syndromes. Overlapping clinical features and molecular heterogeneity of these ciliopathies render their diagnoses challenging. In this review, we describe the clinical characteristics of individual organ disease for each ciliopathy and provide natural history data on kidney, liver, retinal disease progression and central nervous system function.
Collapse
Affiliation(s)
- Angela Grochowsky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meral Gunay-Aygun
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Pediatrics and The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Haque MZ, Ortiz PA. Superoxide increases surface NKCC2 in the rat thick ascending limbs via PKC. Am J Physiol Renal Physiol 2019; 317:F99-F106. [PMID: 31091128 DOI: 10.1152/ajprenal.00232.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The apical Na+-K+-2Cl- cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb (TAL). The free radical superoxide ( O2- ) stimulates TAL NaCl absorption by enhancing NKCC2 activity. In contrast, nitric oxide (NO) scavenges O2- and inhibits NKCC2. NKCC2 activity depends on the number of NKCC2 transporters in the TAL apical membrane and its phosphorylation. We hypothesized that O2- stimulates NKCC2 activity by enhancing apical surface NKCC2 expression. We measured surface NKCC2 expression in rat TALs by surface biotinylation and Western blot analysis. Treatment of TALs with O2- produced by exogenous xanthine oxidase (1 mU/ml) and hypoxanthine (500 µM) stimulated surface NKCC2 expression by ~18 ± 5% (P < 0.05). O2- -stimulated surface NKCC2 expression was blocked by the O2- scavenger tempol (50 µM). Scavenging H2O2 with 100 U/ml catalase did not block the stimulatory effect of xanthine oxidase-hypoxanthine (22 ± 8% increase from control, P < 0.05). Inhibition of endogenous NO production with Nω-nitro-l-arginine methyl ester enhanced surface NKCC2 expression by 21 ± 6% and, when added together with xanthine oxidase-hypoxanthine, increased surface NKCC2 by 41 ± 10% (P < 0.05). Scavenging O2- with superoxide dismutase (300 U/ml) decreased this stimulatory effect by 60% (39 ± 4% to 15 ± 10%, P < 0.05). Protein kinase C inhibition with Gö-6976 (100 nM) blocked O2- -stimulated surface NKCC2 expression (P < 0.05). O2- did not affect NKCC2 phosphorylation at Thr96/101 or its upstream kinases STE20/SPS1-related proline/alanine-rich kinase-oxidative stress-responsive kinase 1. We conclude that O2- increases surface NKCC2 expression by stimulating protein kinase C and that this effect is blunted by endogenous NO. O2- -stimulated apical trafficking of NKCC2 may be involved in the enhanced surface NKCC2 expression observed in Dahl salt-sensitive rats.
Collapse
Affiliation(s)
- Mohammed Ziaul Haque
- Department of Internal Medicine, Hypertension and Vascular Research, Henry Ford Hospital , Detroit, Michigan
| | - Pablo A Ortiz
- Department of Internal Medicine, Hypertension and Vascular Research, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
21
|
Allison SJ. ALMS1-NKCC2 interactions. Nat Rev Nephrol 2018; 15:62. [PMID: 30443014 DOI: 10.1038/s41581-018-0084-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Hearn T. ALMS1 and Alström syndrome: a recessive form of metabolic, neurosensory and cardiac deficits. J Mol Med (Berl) 2018; 97:1-17. [PMID: 30421101 PMCID: PMC6327082 DOI: 10.1007/s00109-018-1714-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/25/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Alström syndrome (AS) is characterised by metabolic deficits, retinal dystrophy, sensorineural hearing loss, dilated cardiomyopathy and multi-organ fibrosis. Elucidating the function of the mutated gene, ALMS1, is critical for the development of specific treatments and may uncover pathways relevant to a range of other disorders including common forms of obesity and type 2 diabetes. Interest in ALMS1 is heightened by the recent discovery of its involvement in neonatal cardiomyocyte cell cycle arrest, a process with potential relevance to regenerative medicine. ALMS1 encodes a ~ 0.5 megadalton protein that localises to the base of centrioles. Some studies have suggested a role for this protein in maintaining centriole-nucleated sensory organelles termed primary cilia, and AS is now considered to belong to the growing class of human genetic disorders linked to ciliary dysfunction (ciliopathies). However, mechanistic details are lacking, and recent studies have implicated ALMS1 in several processes including endosomal trafficking, actin organisation, maintenance of centrosome cohesion and transcription. In line with a more complex picture, multiple isoforms of the protein likely exist and non-centrosomal sites of localisation have been reported. This review outlines the evidence for both ciliary and extra-ciliary functions of ALMS1.
Collapse
Affiliation(s)
- Tom Hearn
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|