Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters








Publication year range
1.
Mol Genet Metab ; 143(1-2): 108575, 2024.
Article in English | MEDLINE | ID: mdl-39243575

ABSTRACT

Alström syndrome (AS) is an inherited rare ciliopathy characterised by multi-organ dysfunction and premature cardiovascular disease. This may manifest as an infantile-onset dilated cardiomyopathy with significant associated mortality. An adult-onset restrictive cardiomyopathy may also feature later in life. Loss of function pathogenic variants in ALMS1 have been identified in AS patients, leading to a lack of ALMS1 protein. The biological role of ALMS1 is unknown, particularly in a cardiovascular context. To understand the role of ALMS1 in infantile cardiomyopathy, the reduction of ALMS1 protein seen in AS patients was modelled using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), in which ALMS1 was knocked out. MuscleMotion analysis and calcium optical mapping experiments suggest that ALMS1 knockout (KO) cells have increased contractility, with altered calcium extrusion and impaired calcium handling dynamics compared to wildtype (WT) counterparts. Seahorse metabolic assays showed ALMS1 knockout iPSC-CMs had increased glycolytic and mitochondrial respiration rates, with ALMS1 knockout cells portraying increased energetic demand and respiratory capacity than WT counterparts. Using senescence associated ß-galactosidase (SA-ß gal) staining assay, we identified increased senescence of ALMS1 knockout iPSC-CMs. Overall, this study provides insights into the molecular mechanisms in AS, particularly the role of ALMS1 in infantile cardiomyopathy in AS, using iPSC-CMs as a 'disease in a dish' model to provide insights into multiple aspects of this complex disease.


Subject(s)
Alstrom Syndrome , Cardiomyopathies , Cell Cycle Proteins , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Induced Pluripotent Stem Cells/metabolism , Alstrom Syndrome/genetics , Alstrom Syndrome/pathology , Alstrom Syndrome/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Calcium/metabolism , Gene Knockout Techniques , Infant , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology
2.
Int J Mol Sci ; 25(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39126056

ABSTRACT

Obesity-related ciliopathies, as a group of ciliopathies including Alström Syndrome and Bardet-Biedl Syndrome, exhibit distinct genetic and phenotypic variability. The understanding of these diseases is highly significant for understanding the functions of primary cilia in the human body, particularly regarding the relationship between obesity and primary cilia. The diagnosis of these diseases primarily relies on clinical presentation and genetic testing. However, there is a significant lack of research on biomarkers to elucidate the variability in clinical manifestations, disease progression, prognosis, and treatment responses. Through an extensive literature review, the paper focuses on obesity-related ciliopathies, reviewing the advancements in the field and highlighting the potential roles of biomarkers in the clinical presentation, diagnosis, and prognosis of these diseases.


Subject(s)
Bardet-Biedl Syndrome , Biomarkers , Ciliopathies , Obesity , Humans , Obesity/metabolism , Obesity/genetics , Ciliopathies/genetics , Ciliopathies/metabolism , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism , Cilia/metabolism , Cilia/pathology , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Animals
3.
Mol Metab ; 84: 101933, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38583571

ABSTRACT

OBJECTIVE: Alström Syndrome (AS), caused by biallelic ALMS1 mutations, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and fatty liver. Prior studies suggest that hyperphagia is accounted for by loss of ALMS1 function in hypothalamic neurones, whereas disproportionate metabolic complications may be due to impaired adipose tissue expandability. We tested this by comparing the metabolic effects of global and mesenchymal stem cell (MSC)-specific Alms1 knockout. METHODS: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα-Cre driver was used to abrogate Alms1 function selectively in MSCs and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα+ Alms1-KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. RESULTS: Assessed on 45% fat diet to promote adipose expansion, global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα-cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfrα expression. CONCLUSIONS: Mesenchymal deletion of Alms1 recapitulates metabolic features of AS, including fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. Hyperphagia in females may depend on Alms1 deficiency in oligodendrocyte precursor cells rather than neurones. AS should be regarded as a forme fruste of lipodystrophy.


Subject(s)
Alstrom Syndrome , Mesenchymal Stem Cells , Mice, Knockout , Animals , Mice , Male , Female , Mesenchymal Stem Cells/metabolism , Alstrom Syndrome/metabolism , Alstrom Syndrome/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Insulin Resistance , Fatty Liver/metabolism , Fatty Liver/genetics , Obesity/metabolism , Obesity/genetics , Hyperphagia/metabolism , Hyperphagia/genetics , Adipose Tissue/metabolism , Mice, Inbred C57BL , Body Composition
4.
Mol Cell Proteomics ; 23(1): 100701, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38122899

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-ß 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 É£-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.


Subject(s)
Alstrom Syndrome , Diabetes Mellitus, Type 2 , Humans , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Cell Cycle Proteins/genetics , Microtubule-Associated Proteins/metabolism , Obesity , Tubulin
5.
Biol Direct ; 18(1): 84, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38062477

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-ß 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-ß stimulation. In addition, we studied alterations in cross-signalling between the TGF-ß pathway and the AKT pathway in this cell line. RESULTS: We found that ALMS1 depletion affects the TGF-ß 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, ß-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-ß 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.


Subject(s)
Alstrom Syndrome , Diabetes Mellitus, Type 2 , Humans , Lipid Metabolism , Diabetes Mellitus, Type 2/metabolism , Proteome/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteomics , Proto-Oncogene Proteins c-akt , Cell Cycle Proteins/metabolism , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Transforming Growth Factor beta/metabolism , Extracellular Matrix/metabolism
6.
Endocrinology ; 164(3)2023 01 09.
Article in English | MEDLINE | ID: mdl-36702623

ABSTRACT

Alström syndrome (AS) is a rare genetic disease caused by ALMS1 mutations, characterized by short stature, and vision and hearing loss. Patients with AS develop the metabolic syndrome, long-term organ complications, and die prematurely. We explored the association between AS and a shortage of hematopoietic stem/progenitor cells (HSPCs), which is linked to metabolic diseases and predicts diabetic complications. We included patients with AS at a national referral center. We measured HSPCs with flow cytometry at baseline and follow-up. We followed patients up to January 2022 for metabolic worsening and end-organ damage. We evaluated HSPC levels and mobilization as well as bone marrow histology in a murine model of AS. In 23 patients with AS, we found significantly lower circulating HSPCs than in healthy blood donors (-40%; P = .002) and age/sex-matched patients (-25%; P = .022). Longitudinally, HSPCs significantly declined by a further 20% in patients with AS over a median of 36 months (interquartile range 30-44). Patients with AS who displayed metabolic deterioration over 5.3 years had lower levels of HSPCs, both at baseline and at last observation, than those who did not deteriorate. Alms1-mutated mice were obese and insulin resistant and displayed significantly reduced circulating HSPCs, despite no overt hematological abnormality. Contrary to what was observed in diabetic mice, HSPC mobilization and bone marrow structure were unaffected. We found depletion of HSPCs in patients with AS, which was recapitulated in Alms1-mutated mice. Larger and longer studies will be needed to establish HSPCs shortage as a driver of metabolic deterioration leading to end-organ damage in AS.


Subject(s)
Alstrom Syndrome , Diabetes Mellitus, Experimental , Metabolic Syndrome , Animals , Mice , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Diabetes Mellitus, Experimental/metabolism , Models, Genetic , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells
7.
J Mol Med (Berl) ; 99(11): 1623-1638, 2021 11.
Article in English | MEDLINE | ID: mdl-34387706

ABSTRACT

Among neonatal cardiomyopathies, primary endocardial fibroelastosis (pEFE) remains a mysterious disease of the endomyocardium that is poorly genetically characterized, affecting 1/5000 live births and accounting for 25% of the entire pediatric dilated cardiomyopathy (DCM) with a devastating course and grave prognosis. To investigate the potential genetic contribution to pEFE, we performed integrative genomic analysis, using whole exome sequencing (WES) and RNA-seq in a female infant with confirmed pathological diagnosis of pEFE. Within regions of homozygosity in the proband genome, WES analysis revealed novel parent-transmitted homozygous mutations affecting three genes with known roles in cilia assembly or function. Among them, a novel homozygous variant [c.1943delA] of uncertain significance in ALMS1 was prioritized for functional genomic and mechanistic analysis. Loss of function mutations of ALMS1 have been implicated in Alstrom syndrome (AS) [OMIM 203800], a rare recessive ciliopathy that has been associated with cardiomyopathy. The variant of interest results in a frameshift introducing a premature stop codon. RNA-seq of the proband's dermal fibroblasts confirmed the impact of the novel ALMS1 variant on RNA-seq reads and revealed dysregulated cellular signaling and function, including the induction of epithelial mesenchymal transition (EMT) and activation of TGFß signaling. ALMS1 loss enhanced cellular migration in patient fibroblasts as well as neonatal cardiac fibroblasts, while ALMS1-depleted cardiomyocytes exhibited enhanced proliferation activity. Herein, we present the unique pathological features of pEFE compared to DCM and utilize integrated genomic analysis to elucidate the molecular impact of a novel mutation in ALMS1 gene in an AS case. Our report provides insights into pEFE etiology and suggests, for the first time to our knowledge, ciliopathy as a potential underlying mechanism for this poorly understood and incurable form of neonatal cardiomyopathy. KEY MESSAGE: Primary endocardial fibroelastosis (pEFE) is a rare form of neonatal cardiomyopathy that occurs in 1/5000 live births with significant consequences but unknown etiology. Integrated genomics analysis (whole exome sequencing and RNA sequencing) elucidates novel genetic contribution to pEFE etiology. In this case, the cardiac manifestation in Alstrom syndrome is pEFE. To our knowledge, this report provides the first evidence linking ciliopathy to pEFE etiology. Infants with pEFE should be examined for syndromic features of Alstrom syndrome. Our findings lead to a better understanding of the molecular mechanisms of pEFE, paving the way to potential diagnostic and therapeutic applications.


Subject(s)
Alstrom Syndrome , Cardiomyopathies , Ciliopathies , Endocardial Fibroelastosis , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Endocardial Fibroelastosis/genetics , Endocardial Fibroelastosis/metabolism , Endocardial Fibroelastosis/pathology , Epithelial-Mesenchymal Transition , Female , Fibroblasts , Humans , Infant , Mutation , Myocardium/metabolism , Myocardium/pathology , Phenotype , RNA-Seq , Transcriptome
8.
Am J Physiol Cell Physiol ; 321(1): C187-C198, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34106786

ABSTRACT

Ca2+ signaling plays a critical role in the regulation of hepatic metabolism by hormones including insulin. Changes in cytoplasmic Ca2+ regulate synthesis and posttranslational modification of key signaling proteins in the insulin pathways. Emerging evidence suggests that hepatocyte intracellular Ca2+ signaling is altered in lipid-loaded liver cells isolated from obese rodent models. The mechanisms of altered Ca2+-insulin and insulin-Ca2+ signaling pathways in obesity remain poorly understood. Here, we show that the kinetics of insulin-initiated intracellular (initial) Ca2+ release from endoplasmic reticulum is significantly impaired in steatotic hepatocytes from obese Alström syndrome mice. Furthermore, exenatide, a glucagon-like peptide-1 (GLP-1) analog, reversed lipid-induced inhibition of intracellular Ca2+ release kinetics in steatotic hepatocytes, without affecting the total content of intracellular Ca2+ released. Exenatide reversed the lipid-induced inhibition of intracellular Ca2+ release, at least partially, via lipid reduction in hepatocytes, which then restored hormone-regulated cytoplasmic Ca2+ signaling and insulin sensitivity. This data provides additional evidence for the important role of Ca2+ signaling pathways in obesity-associated impaired hepatic lipid homeostasis and insulin signaling. It also highlights a potential advantage of GLP-1 analogs when used to treat type 2 diabetes associated with hepatic steatosis.


Subject(s)
Alstrom Syndrome/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Exenatide/pharmacology , Hypoglycemic Agents/pharmacology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Animals , Blood Glucose/metabolism , Calcium/metabolism , Calcium Signaling , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Fluorescent Dyes/metabolism , Fura-2/metabolism , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Insulin/metabolism , Insulin Resistance , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/metabolism , Obesity/pathology , Palmitic Acid/pharmacology
9.
Diabetes ; 70(2): 364-376, 2021 02.
Article in English | MEDLINE | ID: mdl-32994277

ABSTRACT

Obesity is a major risk factor for insulin resistance (IR) and its attendant complications. The pathogenic mechanisms linking them remain poorly understood, partly due to a lack of intermediary monogenic human phenotypes. Here, we report on a monogenic form of IR-prone obesity, Alström syndrome (ALMS). Twenty-three subjects with monogenic or polygenic obesity underwent hyperinsulinemic-euglycemic clamping with concomitant adipose tissue (AT) microdialysis and an in-depth analysis of subcutaneous AT histology. We have shown a relative AT failure in a monogenic obese cohort, a finding supported by observations in a novel conditional mouse model (Alms flin/flin ) and ALMS1-silenced human primary adipocytes, whereas selective reactivation of ALMS1 gene in AT of an ALMS conditional knockdown mouse model (Alms flin/flin ; Adipo-Cre +/- ) restores systemic insulin sensitivity and glucose tolerance. Hence, we show for the first time the relative AT failure in human obese cohorts to be a major determinant of accelerated IR without evidence of lipodystrophy. These new insights into adipocyte-driven IR may assist development of AT-targeted therapeutic strategies for diabetes.


Subject(s)
Adipose Tissue/metabolism , Alstrom Syndrome/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Adipocytes/metabolism , Alstrom Syndrome/genetics , Animals , Diet, High-Fat , Glucose Clamp Technique , Humans , Insulin Resistance/genetics , Mice , Obesity/genetics , Phenotype
10.
J Mol Med (Berl) ; 97(1): 1-17, 2019 01.
Article in English | MEDLINE | ID: mdl-30421101

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.


Subject(s)
Alstrom Syndrome/metabolism , Cell Cycle Proteins/metabolism , Alstrom Syndrome/genetics , Alstrom Syndrome/pathology , Amino Acid Sequence , Animals , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Gene Expression Regulation , Humans , Protein Interaction Maps , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism
11.
Mol Genet Metab ; 125(1-2): 181-191, 2018 09.
Article in English | MEDLINE | ID: mdl-30064963

ABSTRACT

Alström Syndrome is a ciliopathy associated with obesity, insulin resistance/type 2 diabetes mellitus, cardiomyopathy, retinal degeneration, hearing loss, progressive liver and kidney disease, and normal cognitive function. ALMS1, the protein defective in this disorder, localizes to the cytoskeleton, microtubule organizing center, as well as the centrosomes and ciliary basal bodies and plays roles in formation and maintenance of cilia, cell cycle regulation, and endosomal trafficking. Kidney disease in this disorder has not been well characterized. We performed comprehensive multisystem evaluations on 38 patients. Kidney function decreased progressively; eGFR varied inversely with age (p = 0.002). Eighteen percent met the definition for chronic kidney disease (eGFR < 60 mL/min/1.73 m2 and proteinuria); all were adults with median age of 32.8 (20.6-37.9) years. After adjusting for age, there were no significant associations of kidney dysfunction with type 2 diabetes mellitus, dyslipidemia, hypertension, cardiomyopathy or portal hypertension suggesting that kidney disease in AS is a primary manifestation of the syndrome due to lack of ALMS1 protein. Approximately one-third of patients had hyperechogenicity of the renal parenchyma on imaging. While strict control of type 2 diabetes mellitus may decrease kidney-related morbidity and mortality in Alström syndrome, identification of novel targeted therapies is needed.


Subject(s)
Alstrom Syndrome/genetics , Dyslipidemias/genetics , Obesity/genetics , Proteins/genetics , Adult , Alstrom Syndrome/complications , Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Cardiomyopathies/complications , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Cycle Proteins , Dyslipidemias/complications , Dyslipidemias/metabolism , Dyslipidemias/pathology , Female , Humans , Insulin Resistance/genetics , Kidney/metabolism , Kidney/pathology , Kidney Diseases/complications , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mutation , Obesity/complications , Obesity/metabolism , Obesity/pathology , Retinal Degeneration
12.
Acta Ophthalmol ; 96(4): e445-e454, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29193673

ABSTRACT

PURPOSE: We aim to describe ophthalmic characteristics and systemic findings in a cohort of seven patients with cone-rod retinal dystrophy (CORD) caused by pathogenic variants in the ALMS1 gene. METHODS: Seven patients with Alström syndrome (ALMS) were included in the study. A comprehensive ophthalmological examination was performed, including best-corrected visual acuity (BCVA), a semiautomated kinetic visual field exam, colour vision testing, full-field electroretinography testing according to International Society for Clinical Electrophysiology of Vision (ISCEV) standards, spectral domain optical coherence tomography (SD-OCT) and fundus autofluorescence (FAF) imaging, and slit lamp and dilated fundus examination. DNA samples were analysed using Sanger sequencing or exome sequencing. RESULTS: In our cohort, the ocular phenotype presented with a wide variability in retinal function and disease severity. However, age of symptom onset (i.e. nystagmus and photophobia) was at 6-9 months in all patients. These symptoms mostly mislead to the diagnosis of congenital achromatopsia (ACHM), Leber congenital amaurosis (LCA), isolated CORD or Bardet-Biedl syndrome. The systemic manifestations in our cohort were highly variable. CONCLUSION: In summary, we can report that most of our ALMS patients primarily presented with nystagmus and severe photophobia since early childhood interestingly without night blindness in the absence of systemic symptoms. Only genetic testing analysing both nonsyndromic retinal disease (RD) genes and syndromic ciliopathy genes by comprehensive panel sequencing can result in the correct diagnosis, genetically and clinically, with important implication for the physical health of the individual.


Subject(s)
Alstrom Syndrome/genetics , Cone-Rod Dystrophies/genetics , DNA/genetics , Mutation , Proteins/genetics , Visual Acuity , Visual Fields , Adolescent , Adult , Alstrom Syndrome/diagnosis , Alstrom Syndrome/metabolism , Cell Cycle Proteins , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Electroretinography , Female , Genetic Testing , Humans , Male , Pedigree , Phenotype , Proteins/metabolism , Retina/diagnostic imaging , Retina/physiopathology , Tomography, Optical Coherence , Visual Field Tests , Young Adult
13.
PLoS One ; 9(10): e109540, 2014.
Article in English | MEDLINE | ID: mdl-25299671

ABSTRACT

Dysregulation of signaling pathways in adipose tissue leading to insulin resistance can contribute to the development of obesity-related metabolic disorders. Alström Syndrome, a recessive ciliopathy, caused by mutations in ALMS1, is characterized by progressive metabolic alterations such as childhood obesity, hyperinsulinemia, and type 2 diabetes. Here we investigated the role of Alms1 disruption in AT expansion and insulin responsiveness in a murine model for Alström Syndrome. A gene trap insertion in Alms1 on the insulin sensitive C57BL6/Ei genetic background leads to early hyperinsulinemia and a progressive increase in body weight. At 6 weeks of age, before the onset of the metabolic disease, the mutant mice had enlarged fat depots with hypertrophic adipocytes, but without signs of inflammation. Expression of lipogenic enzymes was increased. Pre-adipocytes isolated from mutant animals demonstrated normal adipogenic differentiation but gave rise to mature adipocytes with reduced insulin-stimulated glucose uptake. Assessment of whole body glucose homeostasis revealed glucose intolerance. Insulin stimulation resulted in proper AKT phosphorylation in adipose tissue. However, the total amount of glucose transporter 4 (SLC4A2) and its translocation to the plasma membrane were reduced in mutant adipose depots compared to wildtype littermates. Alterations in insulin stimulated trafficking of glucose transporter 4 are an early sign of metabolic dysfunction in Alström mutant mice, providing a possible explanation for the reduced glucose uptake and the compensatory hyperinsulinemia. The metabolic signaling deficits either reside downstream or are independent of AKT activation and suggest a role for ALMS1 in GLUT4 trafficking. Alström mutant mice represent an interesting model for the development of metabolic disease in which adipose tissue with a reduced glucose uptake can expand by de novo lipogenesis to an obese state.


Subject(s)
Adipocytes/metabolism , Alstrom Syndrome/genetics , DNA-Binding Proteins/genetics , Hyperinsulinism/genetics , Adipocytes/drug effects , Adipocytes/pathology , Adipogenesis/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Animals , Biological Transport , Body Weight , Cell Cycle Proteins , Chloride-Bicarbonate Antiporters/genetics , Chloride-Bicarbonate Antiporters/metabolism , DNA-Binding Proteins/deficiency , Disease Models, Animal , Gene Expression Regulation , Glucose/metabolism , Glucose Intolerance , Humans , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Primary Cell Culture , Signal Transduction
14.
Eur J Med Genet ; 57(2-3): 71-5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24462884

ABSTRACT

BACKGROUND: Alström syndrome is a recessively inherited condition characterised by severe insulin resistance and metabolic syndrome with progression to type 2 diabetes, hepatic dysfunction and coronary artery disease. The metabolic responses to lifestyle changes in the syndrome have not been reported. CASE REPORTS: We describe the effects on glycaemia of intense cycling in two insulin treated Alström patients with diabetes, and the effects of opposite lifestyle changes over one year in two others. METHODS: After practise and clinical assessment two patients aged 21 and 39 years undertook a 380 km cycle ride over 4 days by tandem. The effects of planned reductions in insulin therapies and increased regular carbohydrate ingestion were monitored by frequent capillary blood glucose measurements. Two siblings aged 22 and 25 years underwent assessment of glycaemia, C-peptide/glucose ratio serum lipids, hepatic function and ultrasound, Enhanced Liver Fibrosis test and measures of insulin resistance. Measurements were repeated one year later after profound lifestyle changes. RESULTS: Aerobic exercise strikingly improved blood glucose control despite reduction in insulin dose and increased carbohydrate intake. Increase in exercise and exclusion of fast foods improved all aspects of the metabolic syndrome and induced remission of diabetes in one sibling. Reduction in exercise and consumption of high energy foods in the other resulted in development of type 2 diabetes, severe metabolic syndrome and fatty liver in the other. CONCLUSIONS: Despite dual sensory loss and genetic basis for insulin resistance, Alström patients can successfully ameliorate the metabolic syndrome with lifestyle changes.


Subject(s)
Alstrom Syndrome/therapy , Diabetes Mellitus, Type 2/therapy , Insulin Resistance , Life Style , Adult , Alstrom Syndrome/diagnosis , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Biomarkers/metabolism , Exercise , Exercise Therapy , Female , Humans , Male , Treatment Outcome , Young Adult
15.
Clin Endocrinol (Oxf) ; 79(4): 529-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23445176

ABSTRACT

INTRODUCTION: Alström syndrome (ALMS) is a rare autosomal recessive monogenic disease included in an emerging class of genetic disorders called 'ciliopathies' and is likely to impact the central nervous system as well as metabolic and endocrine function. Individuals with ALMS present clinical features resembling a growth hormone deficiency (GHD) condition, but thus far no study has specifically investigated this aspect in a large population. MATERIAL AND METHODS: Twenty-three patients with ALMS (age, 1-52 years; 11 males, 12 females) were evaluated for anthropometric parameters (growth charts and standard deviation score (SDS) of height, weight, BMI), GH secretion by growth hormone-releasing hormone + arginine test (GHRH-arg), bone age, and hypothalamic-pituitary magnetic resonance imaging (MRI). A group of 17 healthy subjects served as controls in the GH secretion study. Longitudinal retrospective and prospective data were utilized. RESULTS: The length-for-age measurements from birth to 36 months showed normal growth with most values falling within -0·67 SDS to +1·28 SDS. A progressive decrease in stature-for-age was observed after 10 years of age, with a low final height in almost all ALMS subjects (>16-20 years; mean SDS, -2·22 ± 1·16). The subset of 12 patients with ALMS tested for GHRH-arg showed a significantly shorter stature than age-matched controls (154·7 ± 10·6 cm vs 162·9 ± 4·8 cm, P = 0·009) and a mild increase in BMI (Kg/m(2) ) (27·8 ± 4·8 vs 24·1 ± 2·5, P = 0·007). Peak GH after GHRH-arg was significantly lower in patients with ALMS in comparison with controls (11·9 ± 6·9 µg/l vs 86·1 ± 33·2 µg/l, P < 0·0001). Severe GHD was evident biochemically in 50% of patients with ALMS. The 10 adult ALMS patients with GHD showed a reduced height in comparison with those without GHD (149·7 ± 6·2 cm vs 161·9 ± 9·2 cm, P = 0·04). MRIs of the diencephalic and pituitary regions were normal in 11 of 12 patients. Bone age was advanced in 43% of cases. CONCLUSIONS: Our study shows that 50% of nonobese ALMS patients have an inadequate GH reserve to GHRH-arg and may be functionally GH deficient. The short stature reported in ALMS may be at least partially influenced by impairment of GH secretion.


Subject(s)
Alstrom Syndrome/metabolism , Body Height , Body Weight , Growth Disorders/metabolism , Growth Hormone/deficiency , Adolescent , Adult , Alstrom Syndrome/genetics , Alstrom Syndrome/physiopathology , Body Mass Index , Cell Cycle Proteins , Child , Child, Preschool , Diencephalon/diagnostic imaging , Diencephalon/pathology , Female , Growth Disorders/genetics , Growth Disorders/physiopathology , Growth Hormone/metabolism , Humans , Infant , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged , Mutation , Pituitary Gland/diagnostic imaging , Pituitary Gland/pathology , Proteins/genetics , Radiography , Retrospective Studies , Young Adult
16.
PLoS One ; 7(5): e37925, 2012.
Article in English | MEDLINE | ID: mdl-22693585

ABSTRACT

Alström syndrome (ALMS) is a progressive multi-systemic disorder characterized by cone-rod dystrophy, sensorineural hearing loss, childhood obesity, insulin resistance and cardiac, renal, and hepatic dysfunction. The gene responsible for Alström syndrome, ALMS1, is ubiquitously expressed and has multiple splice variants. The protein encoded by this gene has been implicated in ciliary function, cell cycle control, and intracellular transport. To gain better insight into the pathways through which ALMS1 functions, we carried out a yeast two hybrid (Y2H) screen in several mouse tissue libraries to identify ALMS1 interacting partners. The majority of proteins found to interact with the murine carboxy-terminal end (19/32) of ALMS1 were α-actinin isoforms. Interestingly, several of the identified ALMS1 interacting partners (α-actinin 1, α-actinin 4, myosin Vb, rad50 interacting 1 and huntingtin associated protein1A) have been previously associated with endosome recycling and/or centrosome function. We examined dermal fibroblasts from human subjects bearing a disruption in ALMS1 for defects in the endocytic pathway. Fibroblasts from these patients had a lower uptake of transferrin and reduced clearance of transferrin compared to controls. Antibodies directed against ALMS1 N- and C-terminal epitopes label centrosomes and endosomal structures at the cleavage furrow of dividing MDCK cells, respectively, suggesting isoform-specific cellular functions. Our results suggest a role for ALMS1 variants in the recycling endosome pathway and give us new insights into the pathogenesis of a subset of clinical phenotypes associated with ALMS.


Subject(s)
Actinin/metabolism , Endocytosis , Endosomes/metabolism , Proteins/metabolism , Actinin/chemistry , Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Amino Acid Sequence , Animals , Cell Cycle Proteins , Cell Line , Cilia/metabolism , Cytokinesis , Dogs , Fibroblasts/pathology , Humans , Mice , Molecular Sequence Data , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Proteins/chemistry , Stress Fibers/metabolism , Stress Fibers/pathology , Transferrin/metabolism
17.
Mol Biol Cell ; 22(21): 3919-20, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22039061

ABSTRACT

How did I get to become a cell biologist? Or, more generally, why do things happen the way they do? The answer provided by the philosopher Democritus and later adopted by Jacques Monod is "everything existing in the universe is the fruit of chance and necessity." While I read Monod's book Chance and Necessity as an undergraduate student, little did I appreciate the accuracy of this citation and how much of my scientific trajectory would be guided by chance.


Subject(s)
Cell Biology/history , Acetyltransferases/metabolism , Alstrom Syndrome/metabolism , Awards and Prizes , California , Cell Cycle Proteins , Cilia/metabolism , History, 20th Century , History, 21st Century , Humans , Mitosis , Proteins/metabolism
18.
Hum Mol Genet ; 20(3): 466-81, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21071598

ABSTRACT

Alström Syndrome is a life-threatening disease characterized primarily by numerous metabolic abnormalities, retinal degeneration, cardiomyopathy, kidney and liver disease, and sensorineural hearing loss. The cellular localization of the affected protein, ALMS1, has suggested roles in ciliary function and/or ciliogenesis. We have investigated the role of ALMS1 in the cochlea and the pathogenesis of hearing loss in Alström Syndrome. In neonatal rat organ of Corti, ALMS1 was localized to the basal bodies of hair cells and supporting cells. ALMS1 was also evident at the basal bodies of differentiating fibrocytes and marginal cells in the lateral wall. Centriolar ALMS1 expression was retained into maturity. In Alms1-disrupted mice, which recapitulate the neurosensory deficits of human Alström Syndrome, cochleae displayed several cyto-architectural defects including abnormalities in the shape and orientation of hair cell stereociliary bundles. Developing hair cells were ciliated, suggesting that ciliogenesis was largely normal. In adult mice, in addition to bundle abnormalities, there was an accelerated loss of outer hair cells and the progressive appearance of large lesions in stria vascularis. Although the mice progressively lost distortion product otoacoustic emissions, suggesting defects in outer hair cell amplification, their endocochlear potentials were normal, indicating the strial atrophy did not affect its function. These results identify previously unrecognized cochlear histopathologies associated with this ciliopathy that (i) implicate ALMS1 in planar cell polarity signaling and (ii) suggest that the loss of outer hair cells causes the majority of the hearing loss in Alström Syndrome.


Subject(s)
Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Cochlea/ultrastructure , DNA-Binding Proteins/metabolism , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Hearing Loss/genetics , Hearing Loss/pathology , Alstrom Syndrome/genetics , Animals , Cell Cycle Proteins , Cell Differentiation , Cell Polarity , Centrioles , Cilia/ultrastructure , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique , Hearing Loss/metabolism , Mice , Mice, Knockout , Microscopy, Electron , Organ of Corti/ultrastructure , Rats , Rats, Sprague-Dawley , Signal Transduction , Stria Vascularis/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL