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

Publication year range
1.
Nature ; 589(7843): 608-614, 2021 01.
Article in English | MEDLINE | ID: mdl-33408413

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS or progeria) is typically caused by a dominant-negative C•G-to-T•A mutation (c.1824 C>T; p.G608G) in LMNA, the gene that encodes nuclear lamin A. This mutation causes RNA mis-splicing that produces progerin, a toxic protein that induces rapid ageing and shortens the lifespan of children with progeria to approximately 14 years1-4. Adenine base editors (ABEs) convert targeted A•T base pairs to G•C base pairs with minimal by-products and without requiring double-strand DNA breaks or donor DNA templates5,6. Here we describe the use of an ABE to directly correct the pathogenic HGPS mutation in cultured fibroblasts derived from children with progeria and in a mouse model of HGPS. Lentiviral delivery of the ABE to fibroblasts from children with HGPS resulted in 87-91% correction of the pathogenic allele, mitigation of RNA mis-splicing, reduced levels of progerin and correction of nuclear abnormalities. Unbiased off-target DNA and RNA editing analysis did not detect off-target editing in treated patient-derived fibroblasts. In transgenic mice that are homozygous for the human LMNA c.1824 C>T allele, a single retro-orbital injection of adeno-associated virus 9 (AAV9) encoding the ABE resulted in substantial, durable correction of the pathogenic mutation (around 20-60% across various organs six months after injection), restoration of normal RNA splicing and reduction of progerin protein levels. In vivo base editing rescued the vascular pathology of the mice, preserving vascular smooth muscle cell counts and preventing adventitial fibrosis. A single injection of ABE-expressing AAV9 at postnatal day 14 improved vitality and greatly extended the median lifespan of the mice from 215 to 510 days. These findings demonstrate the potential of in vivo base editing as a possible treatment for HGPS and other genetic diseases by directly correcting their root cause.


Subject(s)
Adenine/metabolism , Gene Editing/methods , Mutation , Progeria/genetics , Progeria/therapy , Alleles , Alternative Splicing , Animals , Aorta/pathology , Base Pairing , Child , DNA/genetics , Disease Models, Animal , Female , Fibroblasts/metabolism , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type A/metabolism , Longevity , Male , Mice , Mice, Transgenic , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Progeria/pathology , RNA/genetics
2.
Proc Natl Acad Sci U S A ; 117(22): 12029-12040, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32404427

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a uniformly fatal condition that is especially prevalent in skin, cardiovascular, and musculoskeletal systems. A wide gap exists between our knowledge of the disease and a promising treatment or cure. The aim of this study was to first characterize the musculoskeletal phenotype of the homozygous G608G BAC-transgenic progeria mouse model, and to determine the phenotype changes of HGPS mice after a five-arm preclinical trial of different treatment combinations with lonafarnib, pravastatin, and zoledronic acid. Microcomputed tomography and CT-based rigidity analyses were performed to assess cortical and trabecular bone structure, density, and rigidity. Bones were loaded to failure with three-point bending to assess strength. Contrast-enhanced µCT imaging of mouse femurs was performed to measure glycosaminoglycan content, thickness, and volume of the femoral head articular cartilage. Advanced glycation end products were assessed with a fluorometric assay. The changes demonstrated in the cortical bone structure, rigidity, stiffness, and modulus of the HGPS G608G mouse model may increase the risk for bending and deformation, which could result in the skeletal dysplasia characteristic of HGPS. Cartilage abnormalities seen in this HGPS model resemble changes observed in the age-matched WT controls, including early loss of glycosaminoglycans, and decreased cartilage thickness and volume. Such changes might mimic prevalent degenerative joint diseases in the elderly. Lonafarnib monotherapy did not improve bone or cartilage parameters, but treatment combinations with pravastatin and zoledronic acid significantly improved bone structure and mechanical properties and cartilage structural parameters, which ameliorate the musculoskeletal phenotype of the disease.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Disease Models, Animal , Lamin Type A/genetics , Progeria , Aging/drug effects , Aging/pathology , Animals , Bone and Bones/drug effects , Bone and Bones/pathology , Cartilage/drug effects , Cartilage/pathology , Femur/drug effects , Femur/pathology , Glycosaminoglycans/analysis , Joints/drug effects , Joints/pathology , Lamin Type A/metabolism , Mice , Mice, Transgenic , Mutation , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Phenotype , Piperidines/therapeutic use , Pravastatin/therapeutic use , Progeria/drug therapy , Progeria/genetics , Protein Processing, Post-Translational/drug effects , Pyridines/therapeutic use , X-Ray Microtomography , Zoledronic Acid/therapeutic use
3.
PLoS Genet ; 15(6): e1008196, 2019 06.
Article in English | MEDLINE | ID: mdl-31173582

ABSTRACT

Covalent intermolecular cross-linking of collagen is essential for tissue stability. Recent studies have demonstrated that cyclophilin B (CypB), an endoplasmic reticulum (ER)-resident peptidyl-prolyl cis-trans isomerase, modulates lysine (Lys) hydroxylation of type I collagen impacting cross-linking chemistry. However, the extent of modulation, the molecular mechanism and the functional outcome in tissues are not well understood. Here, we report that, in CypB null (KO) mouse skin, two unusual collagen cross-links lacking Lys hydroxylation are formed while neither was detected in wild type (WT) or heterozygous (Het) mice. Mass spectrometric analysis of type I collagen showed that none of the telopeptidyl Lys was hydroxylated in KO or WT/Het mice. Hydroxylation of the helical cross-linking Lys residues was almost complete in WT/Het but was markedly diminished in KO. Lys hydroxylation at other sites was also lower in KO but to a lesser extent. A key glycosylation site, α1(I) Lys-87, was underglycosylated while other sites were mostly overglycosylated in KO. Despite these findings, lysyl hydroxylases and glycosyltransferase 25 domain 1 levels were significantly higher in KO than WT/Het. However, the components of ER chaperone complex that positively or negatively regulates lysyl hydroxylase activities were severely reduced or slightly increased, respectively, in KO. The atomic force microscopy-based nanoindentation modulus were significantly lower in KO skin than WT. These data demonstrate that CypB deficiency profoundly affects Lys post-translational modifications of collagen likely by modulating LH chaperone complexes. Together, our study underscores the critical role of CypB in Lys modifications of collagen, cross-linking and mechanical properties of skin.


Subject(s)
Cyclophilins/chemistry , Lysine/chemistry , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/chemistry , Skin/enzymology , Animals , Collagen Type I/biosynthesis , Collagen Type I/genetics , Cyclophilins/genetics , Cyclophilins/ultrastructure , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/enzymology , Glycosylation , Heterozygote , Hydroxylation , Lysine/genetics , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Atomic Force , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Protein Processing, Post-Translational/genetics , Skin/chemistry
4.
EMBO J ; 34(22): 2820-39, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26438723

ABSTRACT

Do different neurodegenerative maladies emanate from the failure of a mutual protein folding mechanism? We have addressed this question by comparing mutational patterns that are linked to the manifestation of distinct neurodegenerative disorders and identified similar neurodegeneration-linked proline substitutions in the prion protein and in presenilin 1 that underlie the development of a prion disorder and of familial Alzheimer's disease (fAD), respectively. These substitutions were found to prevent the endoplasmic reticulum (ER)-resident chaperone, cyclophilin B, from assisting presenilin 1 to fold properly, leading to its aggregation, deposition in the ER, reduction of γ-secretase activity, and impaired mitochondrial distribution and function. Similarly, reduced quantities of the processed, active presenilin 1 were observed in brains of cyclophilin B knockout mice. These discoveries imply that reduced cyclophilin activity contributes to the development of distinct neurodegenerative disorders, propose a novel mechanism for the development of certain fAD cases, and support the emerging theme that this disorder can stem from aberrant presenilin 1 function. This study also points at ER chaperones as targets for the development of counter-neurodegeneration therapies.


Subject(s)
Alzheimer Disease/metabolism , Amino Acid Substitution , Brain/metabolism , Presenilin-1/metabolism , Protein Aggregation, Pathological/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , Cell Line , Mice , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Presenilin-1/genetics , Proline/genetics , Proline/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Folding
5.
PLoS Genet ; 12(7): e1006156, 2016 07.
Article in English | MEDLINE | ID: mdl-27441836

ABSTRACT

Recessive osteogenesis imperfecta (OI) is caused by defects in proteins involved in post-translational interactions with type I collagen. Recently, a novel form of moderately severe OI caused by null mutations in TMEM38B was identified. TMEM38B encodes the ER membrane monovalent cation channel, TRIC-B, proposed to counterbalance IP3R-mediated Ca2+ release from intracellular stores. The molecular mechanisms by which TMEM38B mutations cause OI are unknown. We identified 3 probands with recessive defects in TMEM38B. TRIC-B protein is undetectable in proband fibroblasts and osteoblasts, although reduced TMEM38B transcripts are present. TRIC-B deficiency causes impaired release of ER luminal Ca2+, associated with deficient store-operated calcium entry, although SERCA and IP3R have normal stability. Notably, steady state ER Ca2+ is unchanged in TRIC-B deficiency, supporting a role for TRIC-B in the kinetics of ER calcium depletion and recovery. The disturbed Ca2+ flux causes ER stress and increased BiP, and dysregulates synthesis of proband type I collagen at multiple steps. Collagen helical lysine hydroxylation is reduced, while telopeptide hydroxylation is increased, despite increased LH1 and decreased Ca2+-dependent FKBP65, respectively. Although PDI levels are maintained, procollagen chain assembly is delayed in proband cells. The resulting misfolded collagen is substantially retained in TRIC-B null cells, consistent with a 50-70% reduction in secreted collagen. Lower-stability forms of collagen that elude proteasomal degradation are not incorporated into extracellular matrix, which contains only normal stability collagen, resulting in matrix insufficiency. These data support a role for TRIC-B in intracellular Ca2+ homeostasis, and demonstrate that absence of TMEM38B causes OI by dysregulation of calcium flux kinetics in the ER, impacting multiple collagen-specific chaperones and modifying enzymes.


Subject(s)
Calcium/metabolism , Collagen Type I/biosynthesis , Ion Channels/genetics , Osteogenesis Imperfecta/genetics , Adult , Calcium Signaling , Collagen Type I/metabolism , Consanguinity , DNA Mutational Analysis , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Female , Genes, Recessive , Genetic Association Studies , Genetic Predisposition to Disease , Homeostasis , Humans , Infant , Male , Pedigree , Protein Processing, Post-Translational
6.
J Cell Sci ; 129(19): 3635-3647, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27550517

ABSTRACT

Limited detoxification capacity often directs aggregation-prone, potentially hazardous, misfolded proteins to be deposited in designated cytosolic compartments known as 'aggresomes'. The roles of aggresomes as cellular quality control centers, and the cellular origin of the deposits contained within these structures, remain to be characterized. Here, we utilized the observation that the prion protein (PrP, also known as PRNP) accumulates in aggresomes following the inhibition of folding chaperones, members of the cyclophilin family, to address these questions. We found that misfolded PrP molecules must pass through the endoplasmic reticulum (ER) in order to be deposited in aggresomes, that the Golgi plays no role in this process and that cytosolic PrP species are not deposited in pre-existing aggresomes. Prior to their deposition in the aggresome, PrP molecules lose the ER localization signal and have to acquire a GPI anchor. Our discoveries indicate that PrP aggresomes are cytosolic overflow deposition centers for the ER quality control mechanisms and highlight the importance of these structures for the maintenance of protein homeostasis within the ER.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Prion Proteins/metabolism , Protein Aggregates , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclosporine/pharmacology , Cytosol/drug effects , Endoplasmic Reticulum/drug effects , Glycosylation , Glycosylphosphatidylinositols/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Protein Aggregates/drug effects , Protein Folding/drug effects
7.
J Proteome Res ; 16(8): 2914-2923, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28696707

ABSTRACT

Cyclophilin B (CypB) is an endoplasmic reticulum-resident protein that regulates collagen folding, and also contributes to prolyl 3-hydroxylation (P3H) and lysine (Lys) hydroxylation of collagen. In this study, we characterized dentin type I collagen in CypB null (KO) mice, a model of recessive osteogenesis imperfecta type IX, and compared to those of wild-type (WT) and heterozygous (Het) mice. Mass spectrometric analysis demonstrated that the extent of P3H in KO collagen was significantly diminished compared to WT/Het. Lys hydroxylation in KO was significantly diminished at the helical cross-linking sites, α1/α2(I) Lys-87 and α1(I) Lys-930, leading to a significant increase in the under-hydroxylated cross-links and a decrease in fully hydroxylated cross-links. The extent of glycosylation of hydroxylysine residues was, except α1(I) Lys-87, generally higher in KO than WT/Het. Some of these molecular phenotypes were distinct from other KO tissues reported previously, indicating the dentin-specific control mechanism through CypB. Histological analysis revealed that the width of predentin was greater and irregular, and collagen fibrils were sparse and significantly smaller in KO than WT/Het. These results indicate a critical role of CypB in dentin matrix formation, suggesting a possible association between recessive osteogenesis imperfecta and dentin defects that have not been clinically detected.


Subject(s)
Collagen Type I , Cyclophilins/deficiency , Dentin/ultrastructure , Animals , Collagen Type I/ultrastructure , Cyclophilins/physiology , Dentin/pathology , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Glycosylation , Hydroxylation , Lysine/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Osteogenesis Imperfecta , Procollagen-Proline Dioxygenase/metabolism , Protein Processing, Post-Translational
8.
J Biol Chem ; 291(18): 9501-12, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26934917

ABSTRACT

Covalent intermolecular cross-linking provides collagen fibrils with stability. The cross-linking chemistry is tissue-specific and determined primarily by the state of lysine hydroxylation at specific sites. A recent study on cyclophilin B (CypB) null mice, a model of recessive osteogenesis imperfecta, demonstrated that lysine hydroxylation at the helical cross-linking site of bone type I collagen was diminished in these animals (Cabral, W. A., Perdivara, I., Weis, M., Terajima, M., Blissett, A. R., Chang, W., Perosky, J. E., Makareeva, E. N., Mertz, E. L., Leikin, S., Tomer, K. B., Kozloff, K. M., Eyre, D. R., Yamauchi, M., and Marini, J. C. (2014) PLoS Genet 10, e1004465). However, the extent of decrease appears to be tissue- and molecular site-specific, the mechanism of which is unknown. Here we report that although CypB deficiency resulted in lower lysine hydroxylation in the helical cross-linking sites, it was increased in the telopeptide cross-linking sites in tendon type I collagen. This resulted in a decrease in the lysine aldehyde-derived cross-links but generation of hydroxylysine aldehyde-derived cross-links. The latter were absent from the wild type and heterozygous mice. Glycosylation of hydroxylysine residues was moderately increased in the CypB null tendon. We found that CypB interacted with all lysyl hydroxylase isoforms (isoforms 1-3) and a putative lysyl hydroxylase-2 chaperone, 65-kDa FK506-binding protein. Tendon collagen in CypB null mice showed severe size and organizational abnormalities. The data indicate that CypB modulates collagen cross-linking by differentially affecting lysine hydroxylation in a site-specific manner, possibly via its interaction with lysyl hydroxylases and associated molecules. This study underscores the critical importance of collagen post-translational modifications in connective tissue formation.


Subject(s)
Collagen Type I/chemistry , Lysine/chemistry , Animals , Collagen/chemistry , Cyclophilins/metabolism , Hydroxylation , Tendons/metabolism
9.
PLoS Genet ; 10(6): e1004465, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24968150

ABSTRACT

Cyclophilin B (CyPB), encoded by PPIB, is an ER-resident peptidyl-prolyl cis-trans isomerase (PPIase) that functions independently and as a component of the collagen prolyl 3-hydroxylation complex. CyPB is proposed to be the major PPIase catalyzing the rate-limiting step in collagen folding. Mutations in PPIB cause recessively inherited osteogenesis imperfecta type IX, a moderately severe to lethal bone dysplasia. To investigate the role of CyPB in collagen folding and post-translational modifications, we generated Ppib-/- mice that recapitulate the OI phenotype. Knock-out (KO) mice are small, with reduced femoral areal bone mineral density (aBMD), bone volume per total volume (BV/TV) and mechanical properties, as well as increased femoral brittleness. Ppib transcripts are absent in skin, fibroblasts, femora and calvarial osteoblasts, and CyPB is absent from KO osteoblasts and fibroblasts on western blots. Only residual (2-11%) collagen prolyl 3-hydroxylation is detectable in KO cells and tissues. Collagen folds more slowly in the absence of CyPB, supporting its rate-limiting role in folding. However, treatment of KO cells with cyclosporine A causes further delay in folding, indicating the potential existence of another collagen PPIase. We confirmed and extended the reported role of CyPB in supporting collagen lysyl hydroxylase (LH1) activity. Ppib-/- fibroblast and osteoblast collagen has normal total lysyl hydroxylation, while increased collagen diglycosylation is observed. Liquid chromatography/mass spectrometry (LC/MS) analysis of bone and osteoblast type I collagen revealed site-specific alterations of helical lysine hydroxylation, in particular, significantly reduced hydroxylation of helical crosslinking residue K87. Consequently, underhydroxylated forms of di- and trivalent crosslinks are strikingly increased in KO bone, leading to increased total crosslinks and decreased helical hydroxylysine- to lysine-derived crosslink ratios. The altered crosslink pattern was associated with decreased collagen deposition into matrix in culture, altered fibril structure in tissue, and reduced bone strength. These studies demonstrate novel consequences of the indirect regulatory effect of CyPB on collagen hydroxylation, impacting collagen glycosylation, crosslinking and fibrillogenesis, which contribute to maintaining bone mechanical properties.


Subject(s)
Collagen Type I/genetics , Cyclophilins/genetics , Osteogenesis Imperfecta/genetics , Protein Processing, Post-Translational/genetics , Animals , Collagen/chemistry , Collagen/genetics , Collagen/metabolism , Collagen Type I/chemistry , Collagen Type I/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Genes, Recessive , Humans , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Mutation , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Protein Folding
10.
Nat Genet ; 39(3): 359-65, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17277775

ABSTRACT

A recessive form of severe osteogenesis imperfecta that is not caused by mutations in type I collagen has long been suspected. Mutations in human CRTAP (cartilage-associated protein) causing recessive bone disease have been reported. CRTAP forms a complex with cyclophilin B and prolyl 3-hydroxylase 1, which is encoded by LEPRE1 and hydroxylates one residue in type I collagen, alpha1(I)Pro986. We present the first five cases of a new recessive bone disorder resulting from null LEPRE1 alleles; its phenotype overlaps with lethal/severe osteogenesis imperfecta but has distinctive features. Furthermore, a mutant allele from West Africa, also found in African Americans, occurs in four of five cases. All proband LEPRE1 mutations led to premature termination codons and minimal mRNA and protein. Proband collagen had minimal 3-hydroxylation of alpha1(I)Pro986 but excess lysyl hydroxylation and glycosylation along the collagen helix. Proband collagen secretion was moderately delayed, but total collagen secretion was increased. Prolyl 3-hydroxylase 1 is therefore crucial for bone development and collagen helix formation.


Subject(s)
Bone Diseases, Metabolic/genetics , Genes, Recessive , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Osteogenesis Imperfecta/genetics , Proteoglycans/deficiency , Proteoglycans/genetics , Bone Diseases, Metabolic/pathology , Collagen Type I/metabolism , Female , Humans , Male , Mass Spectrometry , Mutation , Osteogenesis Imperfecta/diagnostic imaging , Osteogenesis Imperfecta/pathology , Phenotype , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Prolyl Hydroxylases , Radiography , Time Factors , Ultrasonography, Prenatal
11.
Hum Mutat ; 34(9): 1279-88, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712425

ABSTRACT

Recessive mutations in FKBP10 at 17q21.2, encoding FKBP65, cause both osteogenesis imperfecta (OI) and Bruck syndrome (OI plus congenital contractures). Contractures are a variable manifestation of null/missense FKBP10 mutations. Kuskokwim syndrome (KS) is an autosomal recessive congenital contracture disorder found among Yup'ik Eskimos. Linkage mapping of KS to chromosome 17q21, together with contractures as a feature of FKBP10 mutations, made FKBP10 a candidate gene. We identified a homozygous three-nucleotide deletion in FKBP10 (c.877_879delTAC) in multiple Kuskokwim pedigrees; 3% of regional controls are carriers. The mutation deletes the highly conserved p.Tyr293 residue in FKBP65's third peptidyl-prolyl cis-trans isomerase domain. FKBP10 transcripts are normal, but mutant FKBP65 is destabilized to a residual 5%. Collagen synthesized by KS fibroblasts has substantially decreased hydroxylation of the telopeptide lysine crucial for collagen cross-linking, with 2%-10% hydroxylation in probands versus 60% in controls. Matrix deposited by KS fibroblasts has marked reduction in maturely cross-linked collagen. KS collagen is disorganized in matrix, and fibrils formed in vitro had subtle loosening of monomer packing. Our results imply that FKBP10 mutations affect collagen indirectly, by ablating FKBP65 support for collagen telopeptide hydroxylation by lysyl hydroxylase 2, thus decreasing collagen cross-links in tendon and bone matrix. FKBP10 mutations may also underlie other arthrogryposis syndromes.


Subject(s)
Arthrogryposis/genetics , Contracture/congenital , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Adult , Chromosomes, Human, Pair 17 , Collagen/metabolism , Female , Fibroblasts/metabolism , Genes, Recessive , Genetic Linkage , Homozygote , Humans , Male , Middle Aged , Mutation, Missense , Pedigree , Phylogeny , Sequence Analysis, DNA
12.
N Engl J Med ; 362(6): 521-8, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20089953

ABSTRACT

Osteogenesis imperfecta is a heritable disorder that causes bone fragility. Mutations in type I collagen result in autosomal dominant osteogenesis imperfecta, whereas mutations in either of two components of the collagen prolyl 3-hydroxylation complex (cartilage-associated protein [CRTAP] and prolyl 3-hydroxylase 1 [P3H1]) cause autosomal recessive osteogenesis imperfecta with rhizomelia (shortening of proximal segments of upper and lower limbs) and delayed collagen folding. We identified two siblings who had recessive osteogenesis imperfecta without rhizomelia. They had a homozygous start-codon mutation in the peptidyl-prolyl isomerase B gene (PPIB), which results in a lack of cyclophilin B (CyPB), the third component of the complex. The proband's collagen had normal collagen folding and normal prolyl 3-hydroxylation, suggesting that CyPB is not the exclusive peptidyl-prolyl cis-trans isomerase that catalyzes the rate-limiting step in collagen folding, as is currently thought.


Subject(s)
Codon, Initiator/genetics , Cyclophilins/deficiency , Cyclophilins/genetics , Mutation , Osteogenesis Imperfecta/genetics , Child , Child, Preschool , Collagen/metabolism , Female , Genes, Recessive , Humans , Male , Osteogenesis Imperfecta/metabolism , Pedigree , Phenotype , Procollagen-Proline Dioxygenase/metabolism , Protein Folding
13.
Aging Cell ; 22(9): e13903, 2023 09.
Article in English | MEDLINE | ID: mdl-37365004

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disorder affecting tissues of mesenchymal origin. Most individuals with HGPS harbor a de novo c.1824C > T (p.G608G) mutation in the gene encoding lamin A (LMNA), which activates a cryptic splice donor site resulting in production of the toxic "progerin" protein. Clinical manifestations include growth deficiency, lipodystrophy, sclerotic dermis, cardiovascular defects, and bone dysplasia. Here we utilized the LmnaG609G knock-in (KI) mouse model of HGPS to further define mechanisms of bone loss associated with normal and premature aging disorders. Newborn skeletal staining of KI mice revealed altered rib cage shape and spinal curvature, and delayed calvarial mineralization with increased craniofacial and mandibular cartilage content. MicroCT analysis and mechanical testing of adult femurs indicated increased fragility associated with reduced bone mass, recapitulating the progressive bone deterioration that occurs in HGPS patients. We investigated mechanisms of bone loss in KI mice at the cellular level in bone cell populations. Formation of wild-type and KI osteoclasts from marrow-derived precursors was inhibited by KI osteoblast-conditioned media in vitro, suggesting a secreted factor(s) responsible for decreased osteoclasts on KI trabecular surfaces in vivo. Cultured KI osteoblasts exhibited abnormal differentiation characterized by reduced deposition and mineralization of extracellular matrix with increased lipid accumulation compared to wild-type, providing a mechanism for altered bone formation. Furthermore, quantitative analyses of KI transcripts confirmed upregulation of adipogenic genes both in vitro and in vivo. Thus, osteoblast phenotypic plasticity, inflammation and altered cellular cross-talk contribute to abnormal bone formation in HGPS mice.


Subject(s)
Aging, Premature , Bone Diseases, Developmental , Progeria , Mice , Animals , Progeria/genetics , Progeria/metabolism , Mutation , Lamin Type A/genetics , Lamin Type A/metabolism , Cell Differentiation
14.
Hum Mutat ; 33(11): 1589-98, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22718341

ABSTRACT

Recessive osteogenesis imperfecta (OI) is caused by defects in genes whose products interact with type I collagen for modification and/or folding. We identified a Palestinian pedigree with moderate and lethal forms of recessive OI caused by mutations in FKBP10 or PPIB, which encode endoplasmic reticulum resident chaperone/isomerases FKBP65 and CyPB, respectively. In one pedigree branch, both parents carry a deletion in PPIB (c.563_566delACAG), causing lethal type IX OI in their two children. In another branch, a child with moderate type XI OI has a homozygous FKBP10 mutation (c.1271_1272delCCinsA). Proband FKBP10 transcripts are 4% of control and FKBP65 protein is absent from proband cells. Proband collagen electrophoresis reveals slight band broadening, compatible with ≈10% over-modification. Normal chain incorporation, helix folding, and collagen T(m) support a minimal general collagen chaperone role for FKBP65. However, there is a dramatic decrease in collagen deposited in culture despite normal collagen secretion. Mass spectrometry reveals absence of hydroxylation of the collagen telopeptide lysine involved in cross-linking, suggesting that FKBP65 is required for lysyl hydroxylase activity or access to type I collagen telopeptide lysines, perhaps through its function as a peptidylprolyl isomerase. Proband collagen to organics ratio in matrix is approximately 30% of normal in Raman spectra. Immunofluorescence shows sparse, disorganized collagen fibrils in proband matrix.


Subject(s)
Collagen/metabolism , Mutation , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Tacrolimus Binding Proteins/deficiency , Tacrolimus Binding Proteins/genetics , Base Sequence , Child , Collagen/chemistry , Consanguinity , Cyclophilins/deficiency , Cyclophilins/genetics , DNA Mutational Analysis , Extracellular Matrix/metabolism , Female , Genes, Recessive , Humans , Infant, Newborn , Male , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Osteogenesis Imperfecta/classification , Osteogenesis Imperfecta/diagnostic imaging , Pakistan , Pedigree , Radiography
15.
J Struct Biol ; 180(3): 428-38, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23041293

ABSTRACT

Type I collagen is the most abundant protein in mammals, and is a vital part of the extracellular matrix for numerous tissues. Despite collagen's importance, little is known about its nanoscale morphology in tissues and how morphology relates to mechanical function. This study probes nanoscale structure and mechanical properties of collagen as a function of disease in native hydrated tendons. Wild type tendon and tendon from the Brtl/+ mouse model of Osteogenesis Imperfecta were investigated. An atomic force microscope (AFM) was used to image and indent minimally-processed collagen fibrils in hydrated and dehydrated conditions. AFM was used because of the ability to keep biological tissues as close to their native in situ conditions as possible. The study demonstrated phenotypic difference in Brtl/+ fibril morphology and mechanics in hydrated tendon which became more compelling upon dehydration. Dried tendons had a significant downward shift in fibril D-periodic spacing versus a shift up in wet tendons. Nanoscale changes in morphology in dry samples were accompanied by significant increases in modulus and adhesion force and decreased indentation depth. A minimal mechanical phenotype existed in hydrated samples, possibly due to water masking structural defects within the diseased fibrils. This study demonstrates that collagen nanoscale morphology and mechanics are impacted in Brtl/+ tendons, and that the phenotype can be modulated by the presence or absence of water. Dehydration causes artifacts in biological samples which require water and this factor must be considered for studies at any length scale in collagen-based tissues, especially when characterizing disease-induced differences.


Subject(s)
Collagen Type I/chemistry , Extracellular Matrix/chemistry , Tendons/chemistry , Water/chemistry , Animals , Biomechanical Phenomena , Elastic Modulus , Mice , Microscopy, Atomic Force , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Stress, Mechanical , Tensile Strength
16.
Hum Mol Genet ; 19(2): 223-34, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19846465

ABSTRACT

Null mutations in cartilage-associated protein (CRTAP) and prolyl 3-hydroxylase 1 (P3H1/LEPRE1) cause types VII and VIII OI, respectively, two novel recessive forms of osteogenesis imperfecta (OI) with severe to lethal bone dysplasia and overmodification of the type I collagen helical region. CRTAP and P3H1 form a complex with cyclophilin B (CyPB) in the endoplasmic reticulum (ER) which 3-hydroxylates the Pro986 residue of alpha1(I) and alpha1(II) collagen chains. We investigated the interaction of complex components in fibroblasts from types VII and VIII OI patients. Both CRTAP and P3H1 are absent or reduced on western blots and by immunofluorescence microscopy in cells containing null mutations in either gene. Levels of LEPRE1 or CRTAP transcripts, however, are normal in CRTAP- or LEPRE1-null cells, respectively. Stable transfection of a CRTAP or LEPRE1 expression construct into cells with null mutations for the transfected cDNA restored both CRTAP and P3H1 protein levels. Normalization of collagen helical modification in transfected CRTAP-null cells demonstrated that the restored proteins functioned effectively as a complex. These data indicate that CRTAP and P3H1 are mutually stabilized in the collagen prolyl 3-hydroxylation complex. CyPB levels were unaffected by mutations in either CRTAP or LEPRE1. Proteasomal inhibitors partially rescue P3H1 protein in CRTAP-null cells. In LEPRE1-null cells, secretion of CRTAP is increased compared with control cells and accounts for 15-20% of the decreased CRTAP detected in cells. Thus, mutual stabilization of P3H1 and CRTAP in the ER collagen modification complex is an underlying mechanism for the overlapping phenotype of types VII and VIII OI.


Subject(s)
Collagen/metabolism , Endoplasmic Reticulum/metabolism , Extracellular Matrix Proteins/metabolism , Membrane Glycoproteins/metabolism , Osteogenesis Imperfecta/metabolism , Proteoglycans/metabolism , Base Sequence , Cells, Cultured , Extracellular Matrix Proteins/genetics , Fibroblasts/metabolism , Hydroxylation , Membrane Glycoproteins/genetics , Molecular Chaperones , Molecular Sequence Data , Mutation , Osteogenesis Imperfecta/genetics , Prolyl Hydroxylases , Protein Binding , Proteoglycans/genetics
17.
Genet Med ; 14(5): 543-51, 2012 May.
Article in English | MEDLINE | ID: mdl-22281939

ABSTRACT

PURPOSE: Deficiency of prolyl 3-hydroxylase 1, encoded by LEPRE1, causes recessive osteogenesis imperfecta (OI). We previously identified a LEPRE1 mutation exclusively in African Americans and contemporary West Africans. We hypothesized that this allele originated in West Africa and was introduced to the Americas with the Atlantic slave trade. We aimed to determine the frequency of carriers for this mutation among African Americans and West Africans, and the mutation origin and age. METHODS: Genomic DNA was screened for the mutation using PCR and restriction digestion, and a custom TaqMan genomic single-nucleotide polymorphism assay. The mutation age was estimated using microsatellites and short tandem repeats spanning 4.2 Mb surrounding LEPRE1 in probands and carriers. RESULTS: Approximately 0.4% (95% confidence interval: 0.22-0.68%) of Mid-Atlantic African Americans carry this mutation, estimating recessive OI in 1/260,000 births in this population. In Nigeria and Ghana, 1.48% (95% confidence interval: 0.95-2.30%) of unrelated individuals are heterozygous carriers, predicting that 1/18,260 births will be affected with recessive OI, equal to the incidence of de novo dominant OI. The mutation was not detected in Africans from surrounding countries. All carriers shared a haplotype of 63-770 Kb, consistent with a single founder for this mutation. Using linkage disequilibrium analysis, the mutation was estimated to have originated between 650 and 900 years before present (1100-1350 CE). CONCLUSION: We identified a West African founder mutation for recessive OI in LEPRE1. Nearly 1.5% of Ghanians and Nigerians are carriers. The estimated age of this allele is consistent with introduction to North America via the Atlantic slave trade (1501-1867 CE).


Subject(s)
Founder Effect , Heterozygote , Membrane Glycoproteins/genetics , Osteogenesis Imperfecta/genetics , Proteoglycans/genetics , Black or African American/genetics , Black People/genetics , Cohort Studies , DNA/blood , Genetic Carrier Screening , Genotyping Techniques , Ghana/epidemiology , Humans , Infant, Newborn , Linkage Disequilibrium/genetics , Mutation , Nigeria/epidemiology , North America/epidemiology , Osteogenesis Imperfecta/epidemiology , Prolyl Hydroxylases
18.
Aging Cell ; 20(9): e13457, 2021 09.
Article in English | MEDLINE | ID: mdl-34453483

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare accelerated aging disorder most notably characterized by cardiovascular disease and premature death from myocardial infarction or stroke. The majority of cases are caused by a de novo single nucleotide mutation in the LMNA gene that activates a cryptic splice donor site, resulting in production of a toxic form of lamin A with a 50 amino acid internal deletion, termed progerin. We previously reported the generation of a transgenic murine model of progeria carrying a human BAC harboring the common mutation, G608G, which in the single-copy state develops features of HGPS that are limited to the vascular system. Here, we report the phenotype of mice bred to carry two copies of the BAC, which more completely recapitulate the phenotypic features of HGPS in skin, adipose, skeletal, and vascular tissues. We further show that genetic reduction of the mechanistic target of rapamycin (mTOR) significantly extends lifespan in these mice, providing a rationale for pharmacologic inhibition of the mTOR pathway in the treatment of HGPS.


Subject(s)
Disease Models, Animal , Longevity , Progeria/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , TOR Serine-Threonine Kinases/genetics
19.
Nat Med ; 27(3): 536-545, 2021 03.
Article in English | MEDLINE | ID: mdl-33707773

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare accelerated aging disorder characterized by premature death from myocardial infarction or stroke. It is caused by de novo single-nucleotide mutations in the LMNA gene that activate a cryptic splice donor site, resulting in the production of a toxic form of lamin A, which is termed progerin. Here we present a potential genetic therapeutic strategy that utilizes antisense peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) to block pathogenic splicing of mutant transcripts. Of several candidates, PPMO SRP-2001 provided the most significant decrease in progerin transcripts in patient fibroblasts. Intravenous delivery of SRP-2001 to a transgenic mouse model of HGPS produced significant reduction of progerin transcripts in the aorta, a particularly critical target tissue in HGPS. Long-term continuous treatment with SRP-2001 yielded a 61.6% increase in lifespan and rescue of vascular smooth muscle cell loss in large arteries. These results provide a rationale for proceeding to human trials.


Subject(s)
Oligonucleotides, Antisense/therapeutic use , Progeria/drug therapy , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Morpholinos/chemistry
20.
Cell Tissue Res ; 339(1): 59-70, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19862557

ABSTRACT

Classical osteogenesis imperfecta (OI) is a dominant genetic disorder of connective tissue caused by mutations in either of the two genes encoding type I collagen, COL1A1 and COL1A2. Recent investigations, however, have generated a new paradigm for OI incorporating many of the prototypical features that distinguish dominant and recessive conditions, within a type I collagen framework. We and others have shown that the long-sought cause of the recessive form of OI, first postulated in the Sillence classification, lies in defects in the genes encoding cartilage-associated protein (CRTAP) or prolyl 3-hydroxylase 1 (P3H1/LEPRE1). Together with cyclophilin B (PPIB), CRTAP and P3H1 comprise the collagen prolyl 3-hydroxylation complex, which catalyzes a specific posttranslational modification of types I, II, and V collagen, and may act as a general chaperone. Patients with mutations in CRTAP or LEPRE1 have a lethal to severe osteochondrodystrophy that overlaps with Sillence types II and III OI but has distinctive features. Infants with recessive OI have white sclerae, undertubulation of the long bones, gracile ribs without beading, and a small to normal head circumference. Those who survive to childhood or the teen years have severe growth deficiency and extreme bone fragility. Most causative mutations result in null alleles, with the absence or severe reduction of gene transcripts and proteins. As expected, 3-hydroxylation of the Pro986 residue is absent or severly reduced, but bone severity and survival length do not correlate with the extent of residual hydroxylation. Surprisingly, the collagen produced by cells with an absence of Pro986 hydroxylation has helical overmodification by lysyl hydroxylase and prolyl 4-hydroxylase, indicating that the folding of the collagen helix has been substantially delayed.


Subject(s)
Extracellular Matrix Proteins/genetics , Membrane Glycoproteins/genetics , Mutation , Osteogenesis Imperfecta/genetics , Protein Folding , Protein Processing, Post-Translational , Proteoglycans/genetics , Animals , Collagen/genetics , Collagen/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cyclophilins/genetics , Cyclophilins/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Hydroxylation , Membrane Glycoproteins/metabolism , Molecular Chaperones , Osteogenesis Imperfecta/metabolism , Prolyl Hydroxylases , Protein Structure, Secondary , Proteoglycans/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL