Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Kidney Int ; 105(4): 799-811, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38096951

ABSTRACT

Sporadic cases of apolipoprotein A-IV medullary amyloidosis have been reported. Here we describe five families found to have autosomal dominant medullary amyloidosis due to two different pathogenic APOA4 variants. A large family with autosomal dominant chronic kidney disease (CKD) and bland urinary sediment underwent whole genome sequencing with identification of a chr11:116692578 G>C (hg19) variant encoding the missense mutation p.L66V of the ApoA4 protein. We identified two other distantly related families from our registry with the same variant and two other distantly related families with a chr11:116693454 C>T (hg19) variant encoding the missense mutation p.D33N. Both mutations are unique to affected families, evolutionarily conserved and predicted to expand the amyloidogenic hotspot in the ApoA4 structure. Clinically affected individuals suffered from CKD with a bland urinary sediment and a mean age for kidney failure of 64.5 years. Genotyping identified 48 genetically affected individuals; 44 individuals had an estimated glomerular filtration rate (eGFR) under 60 ml/min/1.73 m2, including all 25 individuals with kidney failure. Significantly, 11 of 14 genetically unaffected individuals had an eGFR over 60 ml/min/1.73 m2. Fifteen genetically affected individuals presented with higher plasma ApoA4 concentrations. Kidney pathologic specimens from four individuals revealed amyloid deposits limited to the medulla, with the mutated ApoA4 identified by mass-spectrometry as the predominant amyloid constituent in all three available biopsies. Thus, ApoA4 mutations can cause autosomal dominant medullary amyloidosis, with marked amyloid deposition limited to the kidney medulla and presenting with autosomal dominant CKD with a bland urinary sediment. Diagnosis relies on a careful family history, APOA4 sequencing and pathologic studies.


Subject(s)
Amyloidosis , Apolipoproteins A , Nephritis, Interstitial , Renal Insufficiency, Chronic , Humans , Middle Aged , Nephritis, Interstitial/diagnosis , Nephritis, Interstitial/genetics , Nephritis, Interstitial/complications , Mutation , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/complications
2.
Am J Med Genet C Semin Med Genet ; 190(3): 309-324, 2022 09.
Article in English | MEDLINE | ID: mdl-36250282

ABSTRACT

The clinical characteristics of autosomal dominant tubulointerstitial kidney disease (ADTKD) include bland urinary sediment, slowly progressive chronic kidney disease (CKD) with many patients reaching end stage renal disease (ESRD) between age 20 and 70 years, and autosomal dominant inheritance. Due to advances in genetic diagnosis, ADTKD is becoming increasingly recognized as a cause of CKD. Pathogenic variants in UMOD, MUC1, and REN are the most common causes of ADTKD. ADTKD-UMOD is also associated with hyperuricemia and gout. ADTKD-REN often presents in childhood with mild hypotension, CKD, hyperkalemia, acidosis, and anemia. ADTKD-MUC1 patients present only with CKD. This review describes the pathophysiology, genetics, clinical manifestation, and diagnosis for ADTKD, with an emphasis on genetic testing and genetic counseling suggestions for patients.


Subject(s)
Genetic Testing , Renal Insufficiency, Chronic , Humans , Young Adult , Adult , Middle Aged , Aged , Uromodulin/genetics , Mutation
3.
Hum Mutat ; 41(9): 1662-1670, 2020 09.
Article in English | MEDLINE | ID: mdl-32623804

ABSTRACT

Classical homocystinuria (HCU) is an inborn error of metabolism caused by loss of cystathionine ß-synthase (CBS) activity with the concomitant buildup of homocysteine. In knockout (KO) mice, a mouse model of HCU, complete lack of CBS is neonatally lethal. Administration of OT-58, an enzyme therapy for HCU, during the first 5 weeks of life rescued KO mice survival by preventing liver disease. Here, we studied the impact of a long-term uninterrupted OT-58 treatment or its absence beyond the neonatal period on liver pathology and metabolism. Plasma and liver metabolites of KO mice on OT-58 treatment were substantially improved or normalized compared with those receiving vehicle. Increased plasma activities of alanine aminotransferase and aspartate aminotransferase of vehicle-injected KO mice suggested the progression of liver damage with age and lack of treatment. At 3 months of age, liver histology showed no signs of hepatopathy in both vehicle- and OT-58-treated KO mice. However, moderate to severe liver disease, characterized by steatosis, hepatocellular necroses, disorganized endoplasmic reticulum, and swollen mitochondria, developed in 6-month-old vehicle-injected KO mice. KO mice on OT-58 treatment remained asymptomatic and were indistinguishable from age-matched healthy controls. Long-term uninterrupted OT-58 treatment was essential to prevent severe liver disease in the KO mouse model of HCU.


Subject(s)
Enzyme Replacement Therapy , Homocystinuria/drug therapy , Liver Diseases/prevention & control , Animals , Disease Models, Animal , Female , Liver/pathology , Male , Mice , Mice, Knockout
4.
Kidney Int ; 98(6): 1589-1604, 2020 12.
Article in English | MEDLINE | ID: mdl-32750457

ABSTRACT

There have been few clinical or scientific reports of autosomal dominant tubulointerstitial kidney disease due to REN mutations (ADTKD-REN), limiting characterization. To further study this, we formed an international cohort characterizing 111 individuals from 30 families with both clinical and laboratory findings. Sixty-nine individuals had a REN mutation in the signal peptide region (signal group), 27 in the prosegment (prosegment group), and 15 in the mature renin peptide (mature group). Signal group patients were most severely affected, presenting at a mean age of 19.7 years, with the prosegment group presenting at 22.4 years, and the mature group at 37 years. Anemia was present in childhood in 91% in the signal group, 69% prosegment, and none of the mature group. REN signal peptide mutations reduced hydrophobicity of the signal peptide, which is necessary for recognition and translocation across the endoplasmic reticulum, leading to aberrant delivery of preprorenin into the cytoplasm. REN mutations in the prosegment led to deposition of prorenin and renin in the endoplasmic reticulum-Golgi intermediate compartment and decreased prorenin secretion. Mutations in mature renin led to deposition of the mutant prorenin in the endoplasmic reticulum, similar to patients with ADTKD-UMOD, with a rate of progression to end stage kidney disease (63.6 years) that was significantly slower vs. the signal (53.1 years) and prosegment groups (50.8 years) (significant hazard ratio 0.367). Thus, clinical and laboratory studies revealed subtypes of ADTKD-REN that are pathophysiologically, diagnostically, and clinically distinct.


Subject(s)
Anemia , Polycystic Kidney Diseases , Adult , Child , Cohort Studies , Female , Humans , Male , Mutation , Polycystic Kidney Diseases/genetics , Renin/genetics , Young Adult
5.
Am J Hum Genet ; 99(1): 174-87, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27392076

ABSTRACT

Autosomal-dominant tubulo-interstitial kidney disease (ADTKD) encompasses a group of disorders characterized by renal tubular and interstitial abnormalities, leading to slow progressive loss of kidney function requiring dialysis and kidney transplantation. Mutations in UMOD, MUC1, and REN are responsible for many, but not all, cases of ADTKD. We report on two families with ADTKD and congenital anemia accompanied by either intrauterine growth retardation or neutropenia. Ultrasound and kidney biopsy revealed small dysplastic kidneys with cysts and tubular atrophy with secondary glomerular sclerosis, respectively. Exclusion of known ADTKD genes coupled with linkage analysis, whole-exome sequencing, and targeted re-sequencing identified heterozygous missense variants in SEC61A1-c.553A>G (p.Thr185Ala) and c.200T>G (p.Val67Gly)-both affecting functionally important and conserved residues in SEC61. Both transiently expressed SEC6A1A variants are delocalized to the Golgi, a finding confirmed in a renal biopsy from an affected individual. Suppression or CRISPR-mediated deletions of sec61al2 in zebrafish embryos induced convolution defects of the pronephric tubules but not the pronephric ducts, consistent with the tubular atrophy observed in the affected individuals. Human mRNA encoding either of the two pathogenic alleles failed to rescue this phenotype as opposed to a complete rescue by human wild-type mRNA. Taken together, these findings provide a mechanism by which mutations in SEC61A1 lead to an autosomal-dominant syndromic form of progressive chronic kidney disease. We highlight protein translocation defects across the endoplasmic reticulum membrane, the principal role of the SEC61 complex, as a contributory pathogenic mechanism for ADTKD.


Subject(s)
Anemia/genetics , Heterozygote , Kidney Diseases/genetics , Mutation , SEC Translocation Channels/genetics , Adult , Aged , Alleles , Amino Acid Sequence , Animals , Biopsy , Child , Chronic Disease , Disease Progression , Endoplasmic Reticulum/metabolism , Exome/genetics , Female , Fetal Growth Retardation/genetics , Genes, Dominant , Golgi Apparatus/metabolism , Humans , Infant, Newborn , Kidney Diseases/pathology , Male , Middle Aged , Models, Molecular , Mutation, Missense/genetics , Neutropenia/genetics , Pedigree , Phenotype , RNA, Messenger/analysis , RNA, Messenger/genetics , SEC Translocation Channels/chemistry , Syndrome , Young Adult , Zebrafish/embryology , Zebrafish/genetics
6.
J Am Soc Nephrol ; 29(9): 2418-2431, 2018 09.
Article in English | MEDLINE | ID: mdl-29967284

ABSTRACT

BACKGROUND: Autosomal dominant tubulointerstitial kidney disease caused by mucin-1 gene (MUC1) mutations (ADTKD-MUC1) is characterized by progressive kidney failure. Genetic evaluation for ADTKD-MUC1 specifically tests for a cytosine duplication that creates a unique frameshift protein (MUC1fs). Our goal was to develop immunohistochemical methods to detect the MUC1fs created by the cytosine duplication and, possibly, by other similar frameshift mutations and to identify novel MUC1 mutations in individuals with positive immunohistochemical staining for the MUC1fs protein. METHODS: We performed MUC1fs immunostaining on urinary cell smears and various tissues from ADTKD-MUC1-positive and -negative controls as well as in individuals from 37 ADTKD families that were negative for mutations in known ADTKD genes. We used novel analytic methods to identify MUC1 frameshift mutations. RESULTS: After technique refinement, the sensitivity and specificity for MUC1fs immunostaining of urinary cell smears were 94.2% and 88.6%, respectively. Further genetic testing on 17 families with positive MUC1fs immunostaining revealed six families with five novel MUC1 frameshift mutations that all predict production of the identical MUC1fs protein. CONCLUSIONS: We developed a noninvasive immunohistochemical method to detect MUC1fs that, after further validation, may be useful in the future for diagnostic testing. Production of the MUC1fs protein may be central to the pathogenesis of ADTKD-MUC1.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Mucin-1/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Biopsy, Needle , Case-Control Studies , Female , Humans , Immunohistochemistry , Incidence , Male , Mutation/genetics , Pedigree , Polycystic Kidney, Autosomal Dominant/mortality , Prognosis , Registries , Retrospective Studies , Risk Assessment
7.
Glycobiology ; 28(6): 382-391, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29548035

ABSTRACT

Blood group B glycosphingolipids (B-GSLs) are substrates of the lysosomal alpha-galactosidase A (AGAL). Similar to its major substrate-globotriaosylceramide (Gb3Cer)-B-GSLs are not degraded and accumulate in the cells of patients affected by an inherited defect of AGAL activity (Fabry disease-FD).The pancreas is a secretory organ known to have high biosynthesis of blood group GSLs. Herein, we provide a comprehensive overview of the biochemical and structural abnormalities in pancreatic tissue from two male FD patients with blood group B. In both patients, we found major accumulation of a variety of complex B-GSLs carrying predominantly hexa- and hepta-saccharide structures. The subcellular pathology was dominated by deposits containing B-glycoconjugates and autofluorescent ceroid. The contribution of Gb3Cer to the storage was minor. This abnormal storage pattern was specific for the pancreatic acinar epithelial cells. Other pancreatic cell types including those of islets of Langerhans were affected much less or not at all.Altogether, we provide evidence for a key role of B-antigens in the biochemical and morphological pathology of the exocrine pancreas in FD patients with blood group B. We believe that our findings will trigger further studies aimed at assessing the potential pancreatic dysfunction in this disease.


Subject(s)
Fabry Disease/metabolism , Glycosphingolipids/metabolism , Pancreas/metabolism , ABO Blood-Group System/metabolism , Acinar Cells/metabolism , Acinar Cells/ultrastructure , Case-Control Studies , Fabry Disease/blood , Fabry Disease/pathology , Galactose/analysis , Galactose/metabolism , Glycosphingolipids/chemistry , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Male , Middle Aged , Pancreas/ultrastructure
8.
Hum Mol Genet ; 25(18): 4062-4079, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466185

ABSTRACT

The Acadian variant of Fanconi Syndrome refers to a specific condition characterized by generalized proximal tubular dysfunction from birth, slowly progressive chronic kidney disease and pulmonary interstitial fibrosis. This condition occurs only in Acadians, a founder population in Nova Scotia, Canada. The genetic and molecular basis of this disease is unknown. We carried out whole exome and genome sequencing and found that nine affected individuals were homozygous for the ultra-rare non-coding variant chr8:96046914 T > C; rs575462405, whereas 13 healthy siblings were either heterozygotes or lacked the mutant allele. This variant is located in intron 2 of NDUFAF6 (NM_152416.3; c.298-768 T > C), 37 base pairs upstream from an alternative splicing variant in NDUFAF6 chr8:96046951 A > G; rs74395342 (c.298-731 A > G). NDUFAF6 encodes NADH:ubiquinone oxidoreductase complex assembly factor 6, also known as C8ORF38. We found that rs575462405-either alone or in combination with rs74395342-affects splicing and synthesis of NDUFAF6 isoforms. Affected kidney and lung showed specific loss of the mitochondria-located NDUFAF6 isoform and ultrastructural characteristics of mitochondrial dysfunction. Accordingly, affected tissues had defects in mitochondrial respiration and complex I biogenesis that were corrected with NDUFAF6 cDNA transfection. Our results demonstrate that the Acadian variant of Fanconi Syndrome results from mitochondrial respiratory chain complex I deficiency. This information may be used in the diagnosis and prevention of this disease in individuals and families of Acadian descent and broadens the spectrum of the clinical presentation of mitochondrial diseases, respiratory chain defects and defects of complex I specifically.


Subject(s)
Electron Transport Complex I/genetics , Fanconi Syndrome/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Adult , Alleles , Canada , Chromosome Mapping , Exome/genetics , Fanconi Syndrome/pathology , Female , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Kidney/metabolism , Kidney/pathology , Lung/metabolism , Lung/pathology , Male , Middle Aged , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mutation
9.
Am J Kidney Dis ; 71(4): 495-500, 2018 04.
Article in English | MEDLINE | ID: mdl-29217307

ABSTRACT

Mucin 1 kidney disease, previously referred to as medullary cystic kidney disease type 1, is a rare hereditary kidney disease. It is one of several diseases now termed autosomal dominant tubulointerstitial kidney disease, as proposed by a KDIGO (Kidney Disease: Improving Global Outcomes) consensus report in 2014. Autosomal dominant tubulointerstitial kidney diseases share common clinical findings, such as autosomal dominant inheritance, bland urinary sediment, absent to mild proteinuria, and progressive loss of kidney function. Although the pathophysiology of mucin 1 kidney disease is still under investigation, genetic testing has been developed to detect the most well-known mutation, a single cytosine insertion into a string of 7 cytosines in the variable-number tandem repeat (VNTR) region of the MUC-1 gene. With this diagnostic tool, nephrologists can offer genetic counseling to affected families and monitor closely for progression of disease. We report a Hispanic patient with a strong family history of chronic kidney disease who tested positive for the MUC1 mutation.


Subject(s)
DNA/genetics , Kidney/pathology , Mucin-1/genetics , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Adult , Biopsy , DNA Mutational Analysis , Female , Humans , Mucin-1/metabolism , Polycystic Kidney, Autosomal Dominant/diagnosis , Polycystic Kidney, Autosomal Dominant/metabolism , Ultrasonography
10.
FASEB J ; 31(12): 5495-5506, 2017 12.
Article in English | MEDLINE | ID: mdl-28821635

ABSTRACT

Classical homocystinuria (HCU) is an inborn error of sulfur amino acid metabolism caused by deficient activity of cystathionine ß-synthase (CBS), resulting in an accumulation of homocysteine and a concomitant decrease of cystathionine and cysteine in blood and tissues. In mice, the complete lack of CBS is neonatally lethal. In this study, newborn CBS-knockout (KO) mice were treated with recombinant polyethyleneglycolylated human truncated CBS (PEG-CBS). Full survival of the treated KO mice, along with a positive impact on metabolite levels in plasma, liver, brain, and kidneys, was observed. The PEG-CBS treatment prevented an otherwise fatal liver disease characterized by steatosis, death of hepatocytes, and ultrastructural abnormalities of endoplasmic reticulum and mitochondria. Furthermore, treatment of the KO mice for 5 mo maintained the plasma metabolite balance and completely prevented osteoporosis and changes in body composition that characterize both the KO model and human patients. These findings argue that early treatment of patients with HCU with PEG-CBS may prevent clinical symptoms of the disease possibly without the need of dietary protein restriction.-Majtan, T., Hulková, H., Park, I., Krijt, J., Kozich, V., Bublil, E. M., Kraus, J. P. Enzyme replacement prevents neonatal death, liver damage, and osteoporosis in murine homocystinuria.


Subject(s)
Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/therapeutic use , Fatty Liver/prevention & control , Homocystinuria/drug therapy , Homocystinuria/enzymology , Liver Diseases/prevention & control , Osteoporosis/prevention & control , Animals , Body Composition/drug effects , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Fatty Liver/enzymology , Female , Homocystinuria/metabolism , Homocystinuria/pathology , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Liver Diseases/enzymology , Male , Mice , Mice, Knockout , Recombinant Proteins/therapeutic use
11.
Am J Hum Genet ; 92(5): 792-9, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23602711

ABSTRACT

The genetic cause of GAPO syndrome, a condition characterized by growth retardation, alopecia, pseudoanodontia, and progressive visual impairment, has not previously been identified. We studied four ethnically unrelated affected individuals and identified homozygous nonsense mutations (c.262C>T [p.Arg88*] and c.505C>T [p.Arg169*]) or splicing mutations (c.1435-12A>G [p.Gly479Phefs*119]) in ANTXR1, which encodes anthrax toxin receptor 1. The nonsense mutations predictably trigger nonsense-mediated mRNA decay, resulting in the loss of ANTXR1. The transcript with the splicing mutation theoretically encodes a truncated ANTXR1 containing a neopeptide composed of 118 unique amino acids in its C terminus. GAPO syndrome's major phenotypic features, which include dental abnormalities and the accumulation of extracellular matrix, recapitulate those found in Antxr1-mutant mice and point toward an underlying defect in extracellular-matrix regulation. Thus, we propose that mutations affecting ANTXR1 function are responsible for this disease's characteristic generalized defect in extracellular-matrix homeostasis.


Subject(s)
Alopecia/genetics , Anodontia/genetics , Chromosomes, Human, Pair 2/genetics , Extracellular Matrix/genetics , Genetic Predisposition to Disease/genetics , Growth Disorders/genetics , Homeostasis/genetics , Neoplasm Proteins/genetics , Optic Atrophies, Hereditary/genetics , Receptors, Cell Surface/genetics , Alopecia/pathology , Alternative Splicing/genetics , Anodontia/pathology , Base Sequence , Codon, Nonsense/genetics , DNA Primers/genetics , Extracellular Matrix/metabolism , Fibroblasts , Fluorescent Antibody Technique , Gene Frequency , Growth Disorders/pathology , Humans , Male , Microfilament Proteins , Molecular Sequence Data , Optic Atrophies, Hereditary/pathology , Pedigree , RNA Splice Sites/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
12.
Brain ; 138(Pt 2): 336-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25567323

ABSTRACT

Severe progressive neurological paediatric disease mucopolysaccharidosis III type C is caused by mutations in the HGSNAT gene leading to deficiency of acetyl-CoA: α-glucosaminide N-acetyltransferase involved in the lysosomal catabolism of heparan sulphate. To understand the pathophysiology of the disease we generated a mouse model of mucopolysaccharidosis III type C by germline inactivation of the Hgsnat gene. At 6-8 months mice showed hyperactivity, and reduced anxiety. Cognitive memory decline was detected at 10 months and at 12-13 months mice showed signs of unbalanced hesitant walk and urinary retention. Lysosomal accumulation of heparan sulphate was observed in hepatocytes, splenic sinus endothelium, cerebral microglia, liver Kupffer cells, fibroblasts and pericytes. Starting from 5 months, brain neurons showed enlarged, structurally abnormal mitochondria, impaired mitochondrial energy metabolism, and storage of densely packed autofluorescent material, gangliosides, lysozyme, phosphorylated tau, and amyloid-ß. Taken together, our data demonstrate for the first time that deficiency of acetyl-CoA: α-glucosaminide N-acetyltransferase causes lysosomal accumulation of heparan sulphate in microglial cells followed by their activation and cytokine release. They also show mitochondrial dysfunction in the neurons and neuronal loss explaining why mucopolysaccharidosis III type C manifests primarily as a neurodegenerative disease.


Subject(s)
Mitochondrial Diseases/pathology , Mucopolysaccharidosis III/pathology , Neuritis/pathology , Neurodegenerative Diseases/pathology , Acetyltransferases/deficiency , Acetyltransferases/genetics , Animals , Behavior, Animal , Energy Metabolism/physiology , Gangliosides/metabolism , Glycosaminoglycans/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mitochondrial Diseases/etiology , Mucopolysaccharidosis III/complications , Mucopolysaccharidosis III/psychology , Neuritis/etiology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/psychology , Neurologic Examination , Proteostasis Deficiencies/pathology
13.
Anal Bioanal Chem ; 407(8): 2283-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25542581

ABSTRACT

Fabry disease is an X-linked lysosomal storage disease due to deficient α-galactosidase A (α-Gal A) activity and the resultant lysosomal accumulation of globotriaosylceramide (Gb3) and related lipids primarily in blood vessels, kidney, heart, and other organs. The renal distribution of stored glycolipid species in the α-Gal A knockout mouse model was compared to that in mice to assess relative distribution and absolute amounts of accumulated sphingolipid isoforms. Twenty isoforms of five sphingolipid groups were visualized by mass spectrometry imaging (MSI), and their distribution was compared with immunohistochemical (IHC) staining of Gb3, the major stored glycosphingolipid in consecutive tissue sections. Quantitative bulk lipid analysis of tissue sections was assessed by electrospray ionization with tandem mass spectrometry (ESI-MS/MS). In contrast to the findings in wild-type mice, all three analytical techniques (MSI, IHC, and ESI-MS/MS) revealed increases in Gb3 isoforms and ceramide dihexosides (composed mostly of galabiosylceramides), respectively. To our knowledge, this is the first report of the distribution of individual molecular species of Gb3 and galabiosylceramides in kidney sections in Fabry disease mouse. In addition, the spatial distribution of ceramides, ceramide monohexosides, and sphingomyelin forms in renal tissue is presented and discussed in the context of their biosynthesis.


Subject(s)
Fabry Disease/metabolism , Kidney/chemistry , Sphingolipids/metabolism , Animals , Disease Models, Animal , Fabry Disease/enzymology , Fabry Disease/genetics , Humans , Immunochemistry , Kidney/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Sphingolipids/chemistry , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
14.
Am J Hum Genet ; 89(2): 241-52, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21820099

ABSTRACT

Autosomal-dominant adult-onset neuronal ceroid lipofuscinosis (ANCL) is characterized by accumulation of autofluorescent storage material in neural tissues and neurodegeneration and has an age of onset in the third decade of life or later. The genetic and molecular basis of the disease has remained unknown for many years. We carried out linkage mapping, gene-expression analysis, exome sequencing, and candidate-gene sequencing in affected individuals from 20 families and/or individuals with simplex cases; we identified in five individuals one of two disease-causing mutations, c.346_348delCTC and c.344T>G, in DNAJC5 encoding cysteine-string protein alpha (CSPα). These mutations-causing a deletion, p.Leu116del, and an amino acid exchange, p.Leu115Arg, respectively-are located within the cysteine-string domain of the protein and affect both palmitoylation-dependent sorting and the amount of CSPα in neuronal cells. The resulting depletion of functional CSPα might cause in parallel the presynaptic dysfunction and the progressive neurodegeneration observed in affected individuals and lysosomal accumulation of misfolded and proteolysis-resistant proteins in the form of characteristic ceroid deposits in neurons. Our work represents an important step in the genetic dissection of a genetically heterogeneous group of ANCLs. It also confirms a neuroprotective role for CSPα in humans and demonstrates the need for detailed investigation of CSPα in the neuronal ceroid lipofuscinoses and other neurodegenerative diseases presenting with neuronal protein aggregation.


Subject(s)
Genes, Dominant/genetics , HSP40 Heat-Shock Proteins/genetics , Membrane Proteins/genetics , Mutation/genetics , Neuronal Ceroid-Lipofuscinoses/epidemiology , Neuronal Ceroid-Lipofuscinoses/genetics , Adult , Age of Onset , Base Sequence , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Chromosome Segregation/genetics , Exons/genetics , Family , Female , Gene Dosage/genetics , Gene Expression Regulation , Genetic Linkage , Humans , Lipoylation , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Molecular Sequence Data , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Pedigree , Protein Transport , Sequence Analysis, DNA
15.
Virchows Arch ; 484(1): 135-140, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37787787

ABSTRACT

Despite the adenoids are regularly removed in patients with mucopolysaccharidoses (MPS), the underlying tissue and cellular pathologies remain understudied. We characterized an (immuno)histopathologic and ultrastructural phenotype dominated by lysosomal storage changes in a specific subset of adenotonsillar paracortical cells in 8 MPS patients (3 MPS I, 3 MPS II, and 2 MPS IIIA). These abnormal cells were effectively detected by an antibody targeting the lysosomal membrane tetraspanin CD63. Important, CD63+ storage vacuoles in these cells lacked the monocytes/macrophages lysosomal marker CD68. Such a distinct patterning of CD63 and CD68 was not present in a patient with infantile neurovisceral variant of acid sphingomyelinase deficiency. The CD63+ storage pathology was absent in two MPS I patients who either received enzyme-replacement therapy or underwent hematopoietic stem cells transplantation prior the adenoidectomy. Our study demonstrates novel features of lysosomal storage patterning and suggests diagnostic utility of CD63 detection in adenotonsillar lymphoid tissue of MPS patients.


Subject(s)
Mucopolysaccharidoses , Humans , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/drug therapy , Mucopolysaccharidoses/genetics , Lymphoid Tissue/pathology , Lysosomes , Enzyme Replacement Therapy , Tetraspanin 30
16.
Kidney Int Rep ; 9(7): 2209-2226, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39081747

ABSTRACT

Introduction: Monoallelic variants in the ALG5 gene encoding asparagine-linked glycosylation protein 5 homolog (ALG5) have been recently shown to disrupt polycystin-1 (PC1) maturation and trafficking via underglycosylation, causing an autosomal dominant polycystic kidney disease-like (ADPKD-like) phenotype and interstitial fibrosis. In this report, we present clinical, genetic, histopathologic, and protein structure and functional correlates of a new ALG5 variant, p.R79W, that we identified in 2 distant genetically related Irish families displaying an atypical late-onset ADPKD phenotype combined with tubulointerstitial damage. Methods: Whole exome and targeted sequencing were used for segregation analysis of available relatives. This was followed by immunohistochemistry examinations of kidney biopsies, and targeted (UMOD, MUC1) and untargeted plasma proteome and N-glycomic studies. Results: We identified a monoallelic ALG5 variant [GRCh37 (NM_013338.5): g.37569565G>A, c.235C>T; p.R79W] that cosegregates in 23 individuals, of whom 18 were clinically affected. We detected abnormal localization of ALG5 in the Golgi apparatus of renal tubular cells in patients' kidney specimens. Further, we detected the pathological accumulation of uromodulin, an N-glycosylated glycosylphosphatidylinositol (GPI)-anchored protein, in the endoplasmic reticulum (ER), but not mucin-1, an O- and N-glycosylated protein. Biochemical investigation revealed decreased plasma and urinary uromodulin levels in clinically affected individuals. Proteomic and glycoproteomic profiling revealed the dysregulation of chronic kidney disease (CKD)-associated proteins. Conclusion: ALG5 dysfunction adversely affects maturation and trafficking of N-glycosylated and GPI anchored protein uromodulin, leading to structural and functional changes in the kidney. Our findings confirm ALG5 as a cause of late-onset ADPKD and provide additional insight into the molecular mechanisms of ADPKD-ALG5.

17.
J Hepatol ; 58(6): 1230-43, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23485521

ABSTRACT

Cholesteryl ester storage disease (CESD) is caused by deficient lysosomal acid lipase (LAL) activity, predominantly resulting in cholesteryl ester (CE) accumulation, particularly in the liver, spleen, and macrophages throughout the body. The disease is characterized by microvesicular steatosis leading to liver failure, accelerated atherosclerosis and premature demise. Although CESD is rare, it is likely that many patients are unrecognized or misdiagnosed. Here, the findings in 135 CESD patients described in the literature are reviewed. Diagnoses were based on liver biopsies, LAL deficiency and/or LAL gene (LIPA) mutations. Hepatomegaly was present in 99.3% of patients; 74% also had splenomegaly. When reported, most patients had elevated serum total cholesterol, LDL-cholesterol, triglycerides, and transaminases (AST, ALT, or both), while HDL-cholesterol was decreased. All 112 liver biopsied patients had the characteristic pathology, which is progressive, and includes microvesicular steatosis, which leads to fibrosis, micronodular cirrhosis, and ultimately to liver failure. Pathognomonic birefringent CE crystals or their remnant clefts were observed in hepatic cells. Extrahepatic manifestations included portal hypertension, esophageal varices, and accelerated atherosclerosis. Liver failure in 17 reported patients resulted in liver transplantation and/or death. Genotyping identified 31 LIPA mutations in 55 patients; 61% of mutations were the common exon 8 splice-junction mutation (E8SJM(-1G>A)), for which 18 patients were homozygous. Genotype/phenotype correlations were limited; however, E8SJM(-1G>A) homozygotes typically had early-onset, slowly progressive disease. Supportive treatment included cholestyramine, statins, and, ultimately, liver transplantation. Recombinant LAL replacement was shown to be effective in animal models, and recently, a phase I/II clinical trial demonstrated its safety and indicated its potential metabolic efficacy.


Subject(s)
Cholesterol Ester Storage Disease/therapy , Cholesterol/blood , Cholesterol Ester Storage Disease/complications , Cholesterol Ester Storage Disease/diagnosis , Cholesterol Ester Storage Disease/genetics , Cholesterol Ester Storage Disease/pathology , Enzyme Replacement Therapy , Humans , Liver/pathology , Liver Transplantation , Triglycerides/blood , Wolman Disease/complications , Wolman Disease
18.
Glycobiology ; 22(5): 725-32, 2012 May.
Article in English | MEDLINE | ID: mdl-22319058

ABSTRACT

A series of six full-term placentas and umbilical cords were examined using the in situ detection of globotriaosylceramide (Gb3Cer), GM1 ganglioside (GM1), GM3 ganglioside (GM3), cholesterol and caveolin 1. Immunohistochemical study showed uniform distinct staining of the apical membrane of villous capillary endothelial cells for Gb3Cer, GM1, GM3 and cholesterol. There was also a strong signal for caveolin 1. The immunophenotype suggests the presence of caveola-associated raft microdomains. The immunophenotype was almost completely shared with the extravillous intravascular trophoblast in the basal plate. It was absent in the endothelial cells of umbilical vessels and in the capillaries of somatic structures (heart, lung, skeletal muscle and skin) in neonates as well as in adults, including capillaries of the proliferative endometrium. Results of in situ analyses were confirmed by lipid chromatographic analysis of tissue homogenates and by tandem mass spectrometry. Lysosomal Gb3Cer turnover was followed in three placentas including umbilical cords from Fabry disease (α-galactosidase A deficiency). Lysosomal storage was restricted to vascular smooth muscle cells and to endothelial cells of umbilical vessels. Placental villous capillary endothelial cells displaying a strong non-lysosomal staining for Gb3Cer were free of lysosomal storage.


Subject(s)
Capillaries/metabolism , Fabry Disease/metabolism , Glycosphingolipids/metabolism , Placenta/metabolism , Female , Humans , Pregnancy , Tandem Mass Spectrometry
19.
Am J Hum Genet ; 85(2): 204-13, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19664745

ABSTRACT

Through linkage analysis and candidate gene sequencing, we identified three unrelated families with the autosomal-dominant inheritance of early onset anemia, hypouricosuric hyperuricemia, progressive kidney failure, and mutations resulting either in the deletion (p.Leu16del) or the amino acid exchange (p.Leu16Arg) of a single leucine residue in the signal sequence of renin. Both mutations decrease signal sequence hydrophobicity and are predicted by bioinformatic analyses to damage targeting and cotranslational translocation of preprorenin into the endoplasmic reticulum (ER). Transfection and in vitro studies confirmed that both mutations affect ER translocation and processing of nascent preprorenin, resulting either in reduced (p.Leu16del) or abolished (p.Leu16Arg) prorenin and renin biosynthesis and secretion. Expression of renin and other components of the renin-angiotensin system was decreased accordingly in kidney biopsy specimens from affected individuals. Cells stably expressing the p.Leu16del protein showed activated ER stress, unfolded protein response, and reduced growth rate. It is likely that expression of the mutant proteins has a dominant toxic effect gradually reducing the viability of renin-expressing cells. This alters the intrarenal renin-angiotensin system and the juxtaglomerular apparatus functionality and leads to nephron dropout and progressive kidney failure. Our findings provide insight into the functionality of renin-angiotensin system and stress the importance of renin analysis in families and individuals with early onset hyperuricemia, anemia, and progressive kidney failure.


Subject(s)
Anemia/genetics , Genes, Dominant , Hyperuricemia/genetics , Kidney Failure, Chronic/genetics , Renin/genetics , Adolescent , Adult , Age of Onset , Anemia/metabolism , Cell Line , Child , Child, Preschool , Computer Simulation , Female , Genetic Linkage , Humans , Hyperuricemia/metabolism , Kidney/cytology , Kidney/ultrastructure , Kidney Failure, Chronic/metabolism , Male , Middle Aged , Mutation , Pedigree , Renin/metabolism , Sequence Analysis, DNA , Young Adult
20.
Histopathology ; 60(7): 1107-13, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22621222

ABSTRACT

AIMS: To identify reliable criteria with which to improve the diagnosis of lysosomal acid lipase (LAL) deficiency of the cholesterol ester storage disease (CESD) type in liver biopsies. METHODS AND RESULTS: We analysed a series of 16 liver biopsies of LAL deficiency of the CESD type confirmed by enzyme testing and DNA sequencing. The biopsy appearances were compared with those of biopsies of other causes of fatty liver. A predominantly microvesicular steatosis in CESD patients could not be reliably distinguished from other causes of fatty liver with cytosolic lipid accumulation in fixed paraffin-embedded tissues routinely stained with haematoxylin and eosin. The presence of luminal (cathepsin D) and membrane lysosomal markers [lysosomal-associated membrane protein (LAMP)1, LAMP2, and lysosomal integral membrane protein 2] around the lipid vacuoles facilitated the diagnosis of CESD in fixed paraffin-embedded material. Additional diagnostic clues included autofluorescent detection of ceroid induction in storage macrophages and the absence of lipopigment in hepatocytes. Stored liquid crystals of cholesteryl esters, which are associated with Maltese cross-type birefringence, were best appreciated in unfixed biopsy samples. CONCLUSIONS: The pathological diagnosis of CESD requires a high index of suspicion, and can be rapidly and effectively appreciated at the light microscopy level, even in routine fixed paraffin-embedded liver samples with immuohistochemical staining for lysosomal markers.


Subject(s)
Cholesterol Ester Storage Disease/diagnosis , Cholesterol Ester Storage Disease/pathology , Liver/pathology , Cathepsin D/metabolism , Child , Child, Preschool , Cholesterol Ester Storage Disease/metabolism , Cohort Studies , Diagnosis, Differential , Fatty Liver/diagnosis , Fatty Liver/metabolism , Fatty Liver/pathology , Humans , Infant , Lipid Metabolism , Liver/metabolism , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/metabolism , Non-alcoholic Fatty Liver Disease , Wolman Disease/diagnosis , Wolman Disease/metabolism , Wolman Disease/pathology , Wolman Disease
SELECTION OF CITATIONS
SEARCH DETAIL