Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 108(11): 2195-2204, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34715011

ABSTRACT

Human mitochondrial RNase P (mt-RNase P) is responsible for 5' end processing of mitochondrial precursor tRNAs, a vital step in mitochondrial RNA maturation, and is comprised of three protein subunits: TRMT10C, SDR5C1 (HSD10), and PRORP. Pathogenic variants in TRMT10C and SDR5C1 are associated with distinct recessive or x-linked infantile onset disorders, resulting from defects in mitochondrial RNA processing. We report four unrelated families with multisystem disease associated with bi-allelic variants in PRORP, the metallonuclease subunit of mt-RNase P. Affected individuals presented with variable phenotypes comprising sensorineural hearing loss, primary ovarian insufficiency, developmental delay, and brain white matter changes. Fibroblasts from affected individuals in two families demonstrated decreased steady state levels of PRORP, an accumulation of unprocessed mitochondrial transcripts, and decreased steady state levels of mitochondrial-encoded proteins, which were rescued by introduction of the wild-type PRORP cDNA. In mt-tRNA processing assays performed with recombinant mt-RNase P proteins, the disease-associated variants resulted in diminished mitochondrial tRNA processing. Identification of disease-causing variants in PRORP indicates that pathogenic variants in all three subunits of mt-RNase P can cause mitochondrial dysfunction, each with distinct pleiotropic clinical presentations.


Subject(s)
Alleles , Genetic Pleiotropy , Mitochondria/enzymology , RNA, Mitochondrial/genetics , RNA, Transfer/genetics , Ribonuclease P/genetics , Adult , Female , Humans , Male , Pedigree
2.
Cell Mol Life Sci ; 79(11): 562, 2022 Oct 22.
Article in English | MEDLINE | ID: mdl-36271951

ABSTRACT

Multifunctional proteins are challenging as it can be difficult to confirm pathomechanisms associated with disease-causing genetic variants. The human 17ß-hydroxysteroid dehydrogenase 10 (HSD10) is a moonlighting enzyme with at least two structurally and catalytically unrelated functions. HSD10 disease was originally described as a disorder of isoleucine metabolism, but the clinical manifestations were subsequently shown to be linked to impaired mtDNA transcript processing due to deficient function of HSD10 in the mtRNase P complex. A surprisingly large number of other, mostly enzymatic and potentially clinically relevant functions have been attributed to HSD10. Recently, HSD10 was reported to exhibit phospholipase C-like activity towards cardiolipins (CL), important mitochondrial phospholipids. To assess the physiological role of the proposed CL-cleaving function, we studied CL architectures in living cells and patient fibroblasts in different genetic backgrounds and lipid environments using our well-established LC-MS/MS cardiolipidomic pipeline. These experiments revealed no measurable effect on CLs, indicating that HSD10 does not have a physiologically relevant function towards CL metabolism. Evolutionary constraints could explain the broad range of reported substrates for HSD10 in vitro. The combination of an essential structural with a non-essential enzymatic function in the same protein could direct the evolutionary trajectory towards improvement of the former, thereby increasing the flexibility of the binding pocket, which is consistent with the results presented here.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases , Hydroxysteroid Dehydrogenases , Humans , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Cardiolipins , Chromatography, Liquid , Tandem Mass Spectrometry , DNA, Mitochondrial , Type C Phospholipases
3.
Hum Mutat ; 41(6): 1145-1156, 2020 06.
Article in English | MEDLINE | ID: mdl-32126153

ABSTRACT

Uncovering frequent motives of action by which variants impair 3' splice site (3'ss) recognition and selection is essential to improve our understanding of this complex process. Through several mini-gene experiments, we demonstrate that the pyrimidine (Y) to purine (R) transversion NM_000267.3(NF1):c.1722-11T>G, although expected to weaken the polypyrimidine tract, causes exon skipping primarily by introducing a novel AG in the AG-exclusion zone (AGEZ) between the authentic 3'ss AG and the branch point. Evaluation of 90 additional noncanonical intronic NF1 3'ss mutations confirmed that 63% of all mutations and 89% (49/55) of the single-nucleotide variants upstream of positions -3 interrupt the AGEZ. Of these AGEZ-interrupting mutations, 24/49 lead to exon skipping suggesting that absence of AG in this region is necessary for accurate 3'ss selection already in the initial steps of splicing. The analysis of 91 noncanonical NF1 3'ss mutations also shows that 90% either introduce a novel AG in the AGEZ, cause a Y>R transversion at position -3 or remove ≥2 Ys in the AGEZ. We confirm in a validation cohort that these three motives distinguish spliceogenic from splice-neutral variants with 85% accuracy and, therefore, are generally applicable to select among variants of unknown significance those likely to affect splicing.


Subject(s)
Introns , Neurofibromin 1/genetics , RNA Splice Sites , RNA Splicing , Adult , Alternative Splicing , Base Sequence , Exons , Female , Humans , Mutation , Ribonucleoproteins, Small Nuclear/genetics
4.
J Biol Chem ; 293(33): 12862-12876, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29880640

ABSTRACT

Mitochondrial tRNAs are transcribed as long polycistronic transcripts of precursor tRNAs and undergo posttranscriptional modifications such as endonucleolytic processing and methylation required for their correct structure and function. Among them, 5'-end processing and purine 9 N1-methylation of mitochondrial tRNA are catalyzed by two proteinaceous complexes with overlapping subunit composition. The Mg2+-dependent RNase P complex for 5'-end cleavage comprises the methyltransferase domain-containing protein tRNA methyltransferase 10C, mitochondrial RNase P subunit (TRMT10C/MRPP1), short-chain oxidoreductase hydroxysteroid 17ß-dehydrogenase 10 (HSD17B10/MRPP2), and metallonuclease KIAA0391/MRPP3. An MRPP1-MRPP2 subcomplex also catalyzes the formation of 1-methyladenosine/1-methylguanosine at position 9 using S-adenosyl-l-methionine as methyl donor. However, a lack of structural information has precluded insights into how these complexes methylate and process mitochondrial tRNA. Here, we used a combination of X-ray crystallography, interaction and activity assays, and small angle X-ray scattering (SAXS) to gain structural insight into the two tRNA modification complexes and their components. The MRPP1 N terminus is involved in tRNA binding and monomer-monomer self-interaction, whereas the C-terminal SPOUT fold contains key residues for S-adenosyl-l-methionine binding and N1-methylation. The entirety of MRPP1 interacts with MRPP2 to form the N1-methylation complex, whereas the MRPP1-MRPP2-MRPP3 RNase P complex only assembles in the presence of precursor tRNA. This study proposes low-resolution models of the MRPP1-MRPP2 and MRPP1-MRPP2-MRPP3 complexes that suggest the overall architecture, stoichiometry, and orientation of subunits and tRNA substrates.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/chemistry , Methyltransferases/chemistry , Models, Molecular , Multienzyme Complexes/chemistry , RNA, Mitochondrial/chemistry , RNA, Transfer/chemistry , Ribonuclease P/chemistry , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Crystallography, X-Ray , Humans , Methyltransferases/metabolism , Multienzyme Complexes/metabolism , RNA, Mitochondrial/metabolism , RNA, Transfer/metabolism , Ribonuclease P/metabolism , Scattering, Small Angle
5.
Am J Hum Genet ; 99(5): 1005-1014, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27745832

ABSTRACT

Periodontal Ehlers-Danlos syndrome (pEDS) is an autosomal-dominant disorder characterized by early-onset periodontitis leading to premature loss of teeth, joint hypermobility, and mild skin findings. A locus was mapped to an approximately 5.8 Mb region at 12p13.1 but no candidate gene was identified. In an international consortium we recruited 19 independent families comprising 107 individuals with pEDS to identify the locus, characterize the clinical details in those with defined genetic causes, and try to understand the physiological basis of the condition. In 17 of these families, we identified heterozygous missense or in-frame insertion/deletion mutations in C1R (15 families) or C1S (2 families), contiguous genes in the mapped locus that encode subunits C1r and C1s of the first component of the classical complement pathway. These two proteins form a heterotetramer that then combines with six C1q subunits. Pathogenic variants involve the subunit interfaces or inter-domain hinges of C1r and C1s and are associated with intracellular retention and mild endoplasmic reticulum enlargement. Clinical features of affected individuals in these families include rapidly progressing periodontitis with onset in the teens or childhood, a previously unrecognized lack of attached gingiva, pretibial hyperpigmentation, skin and vascular fragility, easy bruising, and variable musculoskeletal symptoms. Our findings open a connection between the inflammatory classical complement pathway and connective tissue homeostasis.


Subject(s)
Complement C1r/genetics , Complement C1s/genetics , Ehlers-Danlos Syndrome/genetics , Gene Deletion , Mutation, Missense , Periodontitis/genetics , Adolescent , Adult , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 12/genetics , Ehlers-Danlos Syndrome/diagnosis , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Exome , Female , Genetic Loci , Humans , Male , Pedigree , Periodontitis/diagnosis , Protein Conformation , Young Adult
7.
Cell Physiol Biochem ; 46(2): 713-726, 2018.
Article in English | MEDLINE | ID: mdl-29621776

ABSTRACT

BACKGROUND/AIMS: Reactive dicarbonyl compounds, such as methylglyoxal (MG), contribute to diabetic complications. MG-scavenging capacities of carnosine and anserine, which have been shown to mitigate diabetic nephropathy, were evaluated in vitro and in vivo. METHODS: MG-induced cell toxicity was characterized by MTT and MG-H1-formation, scavenging abilities by Western Blot and NMR spectroscopies, cellular carnosine transport by qPCR and microplate luminescence and carnosine concentration by HPLC. RESULTS: In vitro, carnosine and anserine dose-dependently reduced N-carboxyethyl lysine (CEL) and advanced glycation end products (AGEs) formation. NMR studies revealed the formation of oligo/polymeric products of MG catalyzed by carnosine or anserine. MG toxicity (0.3-1 mM) was dose-dependent for podocytes, tubular and mesangial cells whereas low MG levels (0.2 mM) resulted in increased cell viability in podocytes (143±13%, p<0.001) and tubular cells (129±3%, p<0.001). Incubation with carnosine/anserine did not reduce MG-induced toxicity, independent of incubation times and across large ranges of MG to carnosine/anserine ratios. Cellular carnosine uptake was low (<0.1% in 20 hours) and cellular carnosine concentrations remained unaffected. The putative carnosine transporter PHT1 along with the taurine transporter (TauT) was expressed in all cell types while PEPT1, PEPT2 and PHT2, also belonging to the proton-coupled oligopeptide transporter (POT) family, were only expressed in tubular cells. CONCLUSION: While carnosine and anserine catalyze the formation of MG oligo/polymers, the molar ratios required for protection from MG-induced cellular toxicity are not achievable in renal cells. The effect of carnosine in vivo, to mitigate diabetic nephropathy may therefore be independent upon its ability to scavenge MG and/or carnosine is mainly acting extracellularly.


Subject(s)
Carnosine/chemistry , Carnosine/metabolism , Polymers/chemistry , Pyruvaldehyde/chemistry , Animals , Anserine/analysis , Anserine/chemistry , Anserine/metabolism , Carnosine/analysis , Cell Line , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Glutathione/analysis , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/metabolism , Humans , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Oxidative Stress/drug effects , Peptide Transporter 1/genetics , Peptide Transporter 1/metabolism , Podocytes/cytology , Podocytes/drug effects , Podocytes/metabolism , Polymers/metabolism , Pyruvaldehyde/toxicity , Serum Albumin/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Symporters/genetics , Symporters/metabolism
8.
Hum Mol Genet ; 23(13): 3618-28, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24549042

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 10 (HSD10) is multifunctional protein coded by the X-chromosomal HSD17B10 gene. Mutations in this gene cause HSD10 disease characterized by progressive neurological abnormalities and cardiomyopathy. Disease progression and severity of symptoms is unrelated to the protein's dehydrogenase activity. Recently, it was shown that HSD10 is an essential component of mitochondrial Ribonuclease P (RNase P), an enzyme required for mitochondrial tRNA processing, but little is known about the role of HSD10 in RNase P function. RNase P consists of three different proteins MRPP1, MRPP2 (HSD10) and MRPP3, each of which is essential for RNase P function. Here, we show that HSD10 protein levels are significantly reduced in fibroblasts from patients carrying the HSD17B10 mutation p.R130C. A reduction in HSD10 levels was accompanied by a reduction in MRPP1 protein but not MRPP3 protein. In HSD10 knock-down cells, MRPP1 protein content was also reduced, indicating that HSD10 is important for the maintenance of normal MRPP1 protein levels. Ectopic expression of HSD10 partially restored RNA processing in HSD10 knock-down cells and fibroblasts, and also expression of MRPP1 protein was restored to values comparable to controls. In both, patient fibroblasts and HSD10 knock-down cells, there was evidence of impaired processing of precursor tRNA transcripts of the mitochondrial heavy strand but not the light strand compared with controls. Our findings indicate that HSD10 is important for the maintenance of the MRPP1-HSD10 subcomplex of RNase P and that loss of HSD10 causes impaired mitochondrial precursor transcript processing which may explain mitochondrial dysfunction observed in HSD10 disease.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/metabolism , Methyltransferases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Methyltransferases/genetics , Mutation , RNA, Transfer/genetics , Ribonuclease P/genetics , Ribonuclease P/metabolism
9.
Am J Hum Genet ; 90(4): 701-7, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22424600

ABSTRACT

Kohlschütter-Tönz syndrome (KTS) is an autosomal-recessive disease characterized by the combination of epilepsy, psychomotor regression, and amelogenesis imperfecta. The molecular basis has not yet been elucidated. Here, we report that KTS is caused by mutations in ROGDI. Using a combination of autozygosity mapping and exome sequencing, we identified a homozygous frameshift deletion, c.229_230del (p.Leu77Alafs(∗)64), in ROGDI in two affected individuals from a consanguineous family. Molecular studies in two additional KTS-affected individuals from two unrelated Austrian and Swiss families revealed homozygosity for nonsense mutation c.286C>T (p.Gln96(∗)) and compound heterozygosity for the splice-site mutations c.531+5G>C and c.532-2A>T in ROGDI, respectively. The latter mutation was also found to be heterozygous in the mother of the Swiss affected individual in whom KTS was reported for the first time in 1974. ROGDI is highly expressed throughout the brain and other organs, but its function is largely unknown. Possible interactions with DISC1, a protein involved in diverse cytoskeletal functions, have been suggested. Our finding that ROGDI mutations cause KTS indicates that the protein product of this gene plays an important role in neuronal development as well as amelogenesis.


Subject(s)
Amelogenesis Imperfecta/genetics , Dementia/genetics , Epilepsy/genetics , Membrane Proteins/genetics , Mutation , Nuclear Proteins/genetics , Base Sequence , Chromosome Mapping , Exome , Exons , Female , Heterozygote , Homozygote , Humans , Infant , Male , Molecular Sequence Data
10.
Am J Hum Genet ; 90(2): 201-16, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22265013

ABSTRACT

We report on an autosomal-recessive variant of Ehlers-Danlos syndrome (EDS) characterized by severe muscle hypotonia at birth, progressive scoliosis, joint hypermobility, hyperelastic skin, myopathy, sensorineural hearing impairment, and normal pyridinoline excretion in urine. Clinically, the disorder shares many features with the kyphoscoliotic type of EDS (EDS VIA) and Ullrich congenital muscular dystrophy. Linkage analysis in a large Tyrolean kindred identified a homozygous frameshift mutation in FKBP14 in two affected individuals. Based on the cardinal clinical characteristics of the disorder, four additional individuals originating from different European countries were identified who carried either homozygous or compound heterozygous mutations in FKBP14. FKBP14 belongs to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases). ER-resident FKBPs have been suggested to act as folding catalysts by accelerating cis-trans isomerization of peptidyl-prolyl bonds and to act occasionally also as chaperones. We demonstrate that FKBP14 is localized in the endoplasmic reticulum (ER) and that deficiency of FKBP14 leads to enlarged ER cisterns in dermal fibroblasts in vivo. Furthermore, indirect immunofluorescence of FKBP14-deficient fibroblasts indicated an altered assembly of the extracellular matrix in vitro. These findings suggest that a disturbance of protein folding in the ER affecting one or more components of the extracellular matrix might cause the generalized connective tissue involvement in this disorder. FKBP14 mutation analysis should be considered in all individuals with apparent kyphoscoliotic type of EDS and normal urinary pyridinoline excretion, in particular in conjunction with sensorineural hearing impairment.


Subject(s)
Abnormalities, Multiple/genetics , Ehlers-Danlos Syndrome/genetics , Frameshift Mutation , Hearing Loss/genetics , Peptidylprolyl Isomerase/genetics , Adolescent , Amino Acids/urine , Child , Child, Preschool , Ehlers-Danlos Syndrome/urine , Endoplasmic Reticulum/genetics , Extracellular Matrix/genetics , Female , Fibroblasts/metabolism , Genetic Variation , Hearing Loss/urine , Heterozygote , Homozygote , Humans , Male , Middle Aged , Phenotype , Protein Folding , cis-trans-Isomerases/genetics
11.
Amino Acids ; 47(11): 2367-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26081982

ABSTRACT

Carnosinase 1 (CN1) contributes to diabetic nephropathy by cleaving histidine-dipeptides which scavenge reactive oxygen and carbonyl species and increase nitric oxide (NO) production. In diabetic mice renal CN1 activity is increased, the regulatory mechanisms are unknown. We therefore analysed the in vitro and in vivo regulation of CN1 activity using recombinant and human CN1, and the db/db mouse model of diabetes. Glucose, leptin and insulin did not modify recombinant and human CN1 activity in vitro, glucose did not alter renal CN1 activity of WT or db/db mice ex vivo. Reactive metabolite methylglyoxal and Fenton reagent carbonylated recombinant CN1 and doubled CN1 efficiency. NO S-nitrosylated CN1 and decreased CN1 efficiency for carnosine by 70 % (p < 0.01), but not for anserine. Both CN1 cysteine residues were nitrosylated, the cysteine at position 102 but not at position 229 regulated CN1 activities. In db/db mice, renal CN1 mRNA and protein levels were similar as in non-diabetic controls, CN1 efficiency 1.9 and 1.6 fold higher for carnosine and anserine. Renal carbonyl stress was strongly increased and NO production halved, CN1 highly carbonylated and less S-nitrosylated compared to WT mice. GSH and NO2/3 concentrations were reduced and inversely related with carnosine degradation rate (r = -0.82/-0.85). Thus, reactive metabolites of diabetes upregulate CN1 activity by post-translational modifications, and thus decrease the availability of reactive metabolite-scavenging histidine dipeptides in the kidney in a positive feedback loop. Interference with this vicious circle may represent a new therapeutic target for mitigation of DN.


Subject(s)
Carnosine/metabolism , Diabetes Mellitus/metabolism , Nitric Oxide/metabolism , Pyruvaldehyde/metabolism , Animals , Carnosine/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Dipeptidases/genetics , Dipeptidases/metabolism , Humans , Hydrogen Peroxide/metabolism , Iron/metabolism , Mice , Mice, Mutant Strains , Mutation
12.
World J Surg ; 38(8): 2160-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24682311

ABSTRACT

BACKGROUND: Lipocalin-2 (Lcn-2) is expressed in human neutrophils and epithelial cells, particularly in the presence of inflammation or cancer. It was shown to be highly expressed in various human cancers. Increased protein levels were associated with decreased survival of patients with breast or gastric cancer. The main focus of this work was to analyze the implication of Lcn-2 up-regulation in the genesis of colon cancer. METHODS: Expression of Lcn-2 was analyzed in colorectal carcinoma cell lines, paired colorectal carcinoma tissues, and regular mucosa by Western blot analysis. Lcn-2 immunohistochemical staining was performed in 192 colorectal carcinoma resection specimens and correlated with clinicopathologic parameters. RESULTS: Western blot analysis of colorectal carcinoma tissues demonstrated Lcn-2 overexpression in carcinomas as compared with regular mucosa. Immunohistochemical staining revealed Lcn-2 expression in 179 (93.2%) colorectal carcinoma tissues. Intense immunoreactivity was significantly correlated with metastasis (p = 0.042) and UICC stage (p = 0.027). Survival analysis according to the Kaplan-Meier method revealed a significant association between Lcn-2 overexpressing tumors and overall survival (p < 0.001) and disease-free survival (p < 0.001). CONCLUSIONS: Our data provide evidence that Lcn-2 expression is up-regulated with tumor progression and was found to be a predictor of overall survival.


Subject(s)
Acute-Phase Proteins/analysis , Carcinoma/chemistry , Colorectal Neoplasms/chemistry , Lipocalins/analysis , Proto-Oncogene Proteins/analysis , Acute-Phase Proteins/metabolism , Aged , Carcinoma/mortality , Carcinoma/secondary , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , HT29 Cells , Humans , Intestinal Mucosa/chemistry , Kaplan-Meier Estimate , Lipocalin-2 , Lipocalins/metabolism , Male , Middle Aged , Neoplasm Staging , Proto-Oncogene Proteins/metabolism , Survival Rate , Up-Regulation
13.
NPJ Precis Oncol ; 8(1): 119, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789506

ABSTRACT

Lynch syndrome (LS) and constitutional mismatch repair deficiency (CMMRD) are distinct cancer syndromes caused, respectively, by mono- and bi-allelic germline mismatch repair (MMR) variants. LS predisposes to mainly gastrointestinal and genitourinary cancers in adulthood. CMMRD predisposes to brain, haematological, and LS-spectrum cancers from childhood. Two suspected LS patients with first cancer diagnosis aged 27 or 38 years were found to be homozygous for an MMR (likely) pathogenic variant, MSH6 c.3226C>T (p.(Arg1076Cys)), or variant of uncertain significance (VUS), MLH1 c.306G>A (p.(Glu102=)). MLH1 c.306G>A was shown to cause leaky exon 3 skipping. The apparent genotype-phenotype conflict was resolved by detection of constitutional microsatellite instability in both patients, a hallmark feature of CMMRD. A hypomorphic effect of these and other variants found in additional late onset CMMRD cases, identified by literature review, likely explains a LS-like phenotype. CMMRD testing in carriers of compound heterozygous or homozygous MMR VUS may find similar cases and novel hypomorphic variants. Individualised management of mono- and bi-allelic carriers of hypomorphic MMR variants is needed until we better characterise the associated phenotypes.

14.
Front Immunol ; 14: 1157421, 2023.
Article in English | MEDLINE | ID: mdl-36960056

ABSTRACT

Periodontal Ehlers-Danlos syndrome (pEDS) is an autosomal dominant disorder characterized by early-onset periodontitis leading to premature loss of teeth, lack of attached gingiva and thin and fragile gums leading to gingival recession. Connective tissue abnormalities of pEDS typically include easy bruising, pretibial plaques, distal joint hypermobility, hoarse voice, and less commonly manifestations such as organ or vessel rupture. pEDS is caused by heterozygous missense mutations in C1R and C1S genes of the classical complement C1 complex. Previously we showed that pEDS pathogenic variants trigger intracellular activation of C1r and/or C1s, leading to extracellular presence of activated C1s. However, the molecular link relating activated C1r and C1s proteases to the dysregulated connective tissue homeostasis in pEDS is unknown. Using cell- and molecular-biological assays, we identified activated C1s (aC1s) as an enzyme which degrades collagen I in cell culture and in in vitro assays. Matrix collagen turnover in cell culture was assessed using labelled hybridizing peptides, which revealed fast and comprehensive collagen protein remodeling in patient fibroblasts. Furthermore, collagen I was completely degraded by aC1s when assays were performed at 40°C, indicating that even moderate elevated temperature has a tremendous impact on collagen I integrity. This high turnover is expected to interfere with the formation of a stable ECM and result in tissues with loose compaction a hallmark of the EDS phenotype. Our results indicate that pathogenesis in pEDS is not solely mediated by activation of the complement cascade but by inadequate C1s-mediated degradation of matrix proteins, confirming pEDS as a primary connective tissue disorder.


Subject(s)
Complement C1s , Ehlers-Danlos Syndrome , Humans , Collagen Type I/genetics , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Mutation, Missense , Complement C1s/genetics
15.
Biochim Biophys Acta ; 1813(6): 1144-52, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21406203

ABSTRACT

Mitochondria play central roles in cell life as a source of energy and in cell death by inducing apoptosis. Many important functions of mitochondria change in cancer, and these organelles can be a target of chemotherapy. The widely used anticancer drug doxorubicin (DOX) causes cell death, inhibition of cell cycle/proliferation and mitochondrial impairment. However, the mechanism of such impairment is not completely understood. In our study we used confocal and two-photon fluorescence imaging together with enzymatic and respirometric analysis to study short- and long-term effects of doxorubicin on mitochondria in various human carcinoma cells. We show that short-term (<30 min) effects include i) rapid changes in mitochondrial redox potentials towards a more oxidized state (flavoproteins and NADH), ii) mitochondrial depolarization, iii) elevated matrix calcium levels, and iv) mitochondrial ROS production, demonstrating a complex pattern of mitochondrial alterations. Significant inhibition of mitochondrial endogenous and uncoupled respiration, ATP depletion and changes in the activities of marker enzymes were observed after 48 h of DOX treatment (long-term effects) associated with cell cycle arrest and death.


Subject(s)
Apoptosis/drug effects , Calcium/metabolism , Doxorubicin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Antibiotics, Antineoplastic/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Respiration/drug effects , Cell Survival/drug effects , Citrate (si)-Synthase/metabolism , Dose-Response Relationship, Drug , Electron Transport Complex I/metabolism , HT29 Cells , Humans , Microscopy, Confocal , Mitochondria/metabolism , Mitochondria/physiology , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Time Factors , Uncoupling Agents/pharmacology
16.
Transpl Int ; 23(12): 1282-92, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20561305

ABSTRACT

Chronic rejection (CR) remains an unsolved hurdle for long-term heart transplant survival. The effect of cold ischemia (CI) on progression of CR and the mechanisms resulting in functional deficit were investigated by studying gene expression, mitochondrial function, and enzymatic activity. Allogeneic (Lew→F344) and syngeneic (Lew→Lew) heart transplantations were performed with or without 10 h of CI. After evaluation of myocardial contraction, hearts were excised at 2, 10, 40, and 60 days for investigation of vasculopathy, gene expression, enzymatic activities, and mitochondrial respiration. Gene expression studies identified a gene cluster coding for subunits of the mitochondrial electron transport chain regulated in response to CI and CR. Myocardial performance, mitochondrial function, and mitochondrial marker enzyme activities declined in all allografts with time after transplantation. These declines were more rapid and severe in CI allografts (CR-CI) and correlated well with progression of vasculopathy and fibrosis. Mitochondria related gene expression and mitochondrial function are substantially compromised with the progression of CR and show that CI impacts on progression, gene profile, and mitochondrial function of CR. Monitoring mitochondrial function and enzyme activity might allow for earlier detection of CR and cardiac allograft dysfunction.


Subject(s)
Cold Ischemia , Graft Rejection , Heart Transplantation/physiology , Mitochondria, Heart/physiology , Animals , Gene Expression Profiling , Heart Transplantation/adverse effects , Male , Myocardial Contraction , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Transplantation, Homologous
17.
Cancer Sci ; 100(8): 1414-20, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19493271

ABSTRACT

Dkk-3 is proposed to be a new specific marker for tumor endothelial cells. Here we analyzed the clinical relevance of Dkk-3 expression in pancreas adenocarcinomas and determined its role on endothelial cell growth in vitro. Microvessel density in tumor samples was immunohistochemically determined using Dkk-3 and CD31 as endothelial cell markers, respectively. Based on the median microvessel density as a cut-off point, patients were categorized into high and low microvessel density groups and a correlation with survival and clinical parameters was assessed. Moreover, the role of Dkk-3 expression on chemosensitivity of endothelial cells was analyzed. In contrast to CD31 staining, Dkk-3-positive vessels were found only in tumor tissue and Dkk-3 microvessel density significantly correlated negative with tumor grading. In survival analysis the median survival time was 7 months for patients with Dkk-3 low, and 15 months for Dkk-3 high microvessel density (P = 0.0013). Subset analysis of patients receiving gemcitabine therapy showed that overall survival was significantly decreased in Dkk-3 low tumors than in high tumors (P = 0.009). In Cox regression Dkk-3 emerged as a significant independent parameter (P = 0.024). Dkk-3 overexpression in endothelial cells resulted in significantly enhanced growth inhibition after 5-fluorouracil or gemcitabine treatment compared to control endothelial cells and cancer cell lines. Dkk-3 low microvessel density was associated with tumor progression and worse clinical outcome. Overexpression of Dkk-3 enhanced endothelial cell growth inhibition to chemotherapeutic drugs. Therefore, we suggest that Dkk-3 high microvessel density may help to select patients who may benefit from chemotherapy.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Endothelium/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/pathology , Adaptor Proteins, Signal Transducing , Adenocarcinoma/genetics , Adenoviridae/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cells, Cultured , Chemokines , Endothelium/pathology , Endothelium, Vascular/cytology , Follow-Up Studies , Gene Expression , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Pancreatic Neoplasms/genetics , Prognosis , Retrospective Studies , Survival Analysis , Time Factors , Transfection , Umbilical Veins/cytology
19.
Front Immunol ; 10: 2537, 2019.
Article in English | MEDLINE | ID: mdl-31749804

ABSTRACT

Heterozygous missense or in-frame insertion/deletion mutations in complement 1 subunits C1r and C1s cause periodontal Ehlers-Danlos Syndrome (pEDS), a specific EDS subtype characterized by early severe periodontal destruction and connective tissue abnormalities like easy bruising, pretibial haemosiderotic plaques, and joint hypermobility. We report extensive functional studies of 16 C1R variants associated with pEDS by in-vitro overexpression studies in HEK293T cells followed by western blot, size exclusion chromatography and surface plasmon resonance analyses. Patient-derived skin fibroblasts were analyzed by western blot and Enzyme-linked Immunosorbent Assay (ELISA). Overexpression of C1R variants in HEK293T cells revealed that none of the pEDS variants was integrated into the C1 complex but cause extracellular presence of catalytic C1r/C1s activities. Variants showed domain-specific abnormalities of intracellular processing and secretion with preservation of serine protease function in the supernatant. In contrast to C1r wild type, and with the exception of a C1R missense variant disabling a C1q binding site, pEDS variants had different impact on the cell: retention of C1r fragments inside the cell, secretion of aggregates, or a new C1r cleavage site. Overexpression of C1R variants in HEK293T as well as western blot analyses of patient fibroblasts showed decreased levels of secreted C1r. Importantly, all available patient fibroblasts exhibited activated C1s and activation of externally added C4 in the supernatant while control cell lines secreted proenzyme C1s and showed no increase in C4 activation. The central elements in the pathogenesis of pEDS seem to be the intracellular activation of C1r and/or C1s, and extracellular presence of activated C1s that independently of microbial triggers can activate the classical complement cascade.


Subject(s)
Complement C1/immunology , Complement C1r/immunology , Ehlers-Danlos Syndrome/immunology , Periodontal Diseases/immunology , Cells, Cultured , Complement Activation , Complement C1r/genetics , Ehlers-Danlos Syndrome/genetics , Fibroblasts/immunology , Humans , Mutation , Periodontal Diseases/genetics
20.
Front Immunol ; 10: 2962, 2019.
Article in English | MEDLINE | ID: mdl-31921203

ABSTRACT

Ehlers-Danlos syndromes (EDS) are clinically and genetically heterogeneous disorders characterized by soft connective tissue alteration like joint hypermobility and skin hyper-extensibility. We previously identified heterozygous missense mutations in the C1R and C1S genes, coding for the complement C1 proteases, in patients affected by periodontal EDS, a specific EDS subtype hallmarked by early severe periodontitis leading to premature loss of teeth and connective tissue alterations. Up to now, there is no clear molecular link relating the nominal role of the C1r and C1s proteases, which is to activate the classical complement pathway, to these heterogeneous symptoms of periodontal EDS syndrome. We aim therefore to elucidate the functional effect of these mutations, at the molecular and enzymatic levels. To explore the molecular consequences, a set of cell transfection experiments, recombinant protein purification, mass spectroscopy and N-terminal analyses have been performed. Focusing on the results obtained on two different C1S variants, namely p.Val316del and p.Cys294Arg, we show that HEK293-F cells stably transfected with the corresponding C1s variant plasmids, unexpectedly, do not secrete the full-length mutated C1s, but only a truncated Fg40 fragment of 40 kDa, produced at very low levels. Detailed analyses of the Fg40 fragments purified for the two C1s variants show that they are identical, which was also unexpected. This suggests that local misfolding of the CCP1 module containing the patient mutation exposes a novel cleavage site, between Lys353 and Cys354, which is not normally accessible. The mutation-induced Fg40 fragment contains the intact C-terminal serine protease domain but not the N-terminal domain mediating C1s interaction with the other C1 subunits, C1r, and C1q. Thus, Fg40 enzymatic activity escapes the normal physiological control of C1s activity within C1, potentially providing a loss-of-control. Comparative enzymatic analyses show that Fg40 retains the native esterolytic activity of C1s, as well as its cleavage efficiency toward the ancillary alarmin HMGB1 substrate, for example, whereas the nominal complement C4 activation cleavage is impaired. These new results open the way to further molecular explorations possibly involving subsidiary C1s targets.


Subject(s)
Complement C1r , Complement C1s , Ehlers-Danlos Syndrome , Mutation, Missense , Periodontal Diseases , Amino Acid Substitution , Complement C1r/genetics , Complement C1r/immunology , Complement C1s/genetics , Complement C1s/immunology , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/immunology , Ehlers-Danlos Syndrome/pathology , HEK293 Cells , Humans , Periodontal Diseases/genetics , Periodontal Diseases/immunology , Periodontal Diseases/pathology , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL