scholarly journals Restoration of Osteogenesis by CRISPR/Cas9 Genome Editing of the Mutated COL1A1 Gene in Osteogenesis Imperfecta

2021 ◽  
Vol 10 (14) ◽  
pp. 3141
Author(s):  
Hyerin Jung ◽  
Yeri Alice Rim ◽  
Narae Park ◽  
Yoojun Nam ◽  
Ji Hyeon Ju

Osteogenesis imperfecta (OI) is a genetic disease characterized by bone fragility and repeated fractures. The bone fragility associated with OI is caused by a defect in collagen formation due to mutation of COL1A1 or COL1A2. Current strategies for treating OI are not curative. In this study, we generated induced pluripotent stem cells (iPSCs) from OI patient-derived blood cells harboring a mutation in the COL1A1 gene. Osteoblast (OB) differentiated from OI-iPSCs showed abnormally decreased levels of type I collagen and osteogenic differentiation ability. Gene correction of the COL1A1 gene using CRISPR/Cas9 recovered the decreased type I collagen expression in OBs differentiated from OI-iPSCs. The osteogenic potential of OI-iPSCs was also recovered by the gene correction. This study suggests a new possibility of treatment and in vitro disease modeling using patient-derived iPSCs and gene editing with CRISPR/Cas9.

Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 2165
Author(s):  
Graziana Monaco ◽  
Yann D. Ladner ◽  
Alicia J. El Haj ◽  
Nicholas R. Forsyth ◽  
Mauro Alini ◽  
...  

In the field of tissue engineering, progress has been made towards the development of new treatments for cartilage and bone defects. However, in vitro culture conditions for human bone marrow mesenchymal stromal cells (hBMSCs) have not yet been fully defined. To improve our understanding of cartilage and bone in vitro differentiation, we investigated the effect of culture conditions on hBMSC differentiation. We hypothesized that the use of two different culture media including specific growth factors, TGFβ1 or BMP2, as well as low (2% O2) or high (20% O2) oxygen tension, would improve the chondrogenic and osteogenic potential, respectively. Chondrogenic and osteogenic differentiation of hBMSCs isolated from multiple donors and expanded under the same conditions were directly compared. Chondrogenic groups showed a notable upregulation of chondrogenic markers compared with osteogenic groups. Greater sGAG production and deposition, and collagen type II and I accumulation occurred for chondrogenic groups. Chondrogenesis at 2% O2 significantly reduced ALP gene expression and reduced type I collagen deposition, producing a more stable and less hypertrophic chondrogenic phenotype. An O2 tension of 2% did not inhibit osteogenic differentiation at the protein level but reduced ALP and OC gene expression. An upregulation of ALP and OC occurred during osteogenesis in BMP2 containing media under 20% O2; BMP2 free osteogenic media downregulated ALP and also led to higher sGAG release. A higher mineralization was observed in the presence of BMP2 during osteogenesis. This study demonstrates how the modulation of O2 tension, combined with tissue-specific growth factors and media composition can be tailored in vitro to promote chondral or endochondral differentiation while using the same donor cell population.


1991 ◽  
Vol 87 (1) ◽  
pp. 33-40 ◽  
Author(s):  
Charles J. Pruchno ◽  
Daniel H. Cohn ◽  
Gillian A. Wallis ◽  
Marcia C. Willing ◽  
Barbra J. Starman ◽  
...  

Cells ◽  
2021 ◽  
Vol 11 (1) ◽  
pp. 26
Author(s):  
Giorgia Borciani ◽  
Giorgia Montalbano ◽  
Priscila Melo ◽  
Nicola Baldini ◽  
Gabriela Ciapetti ◽  
...  

Osteoporosis is a worldwide disease resulting in the increase of bone fragility and enhanced fracture risk in adults. In the context of osteoporotic fractures, bone tissue engineering (BTE), i.e., the use of bone substitutes combining biomaterials, cells, and other factors, is considered a potential alternative to conventional treatments. Innovative scaffolds need to be tested in in vitro systems where the simultaneous presence of osteoblasts (OBs) and osteoclasts (OCs), the two main players of bone remodeling, is required to mimic their crosstalk and molecular cooperation. To this aim, two composite materials were developed, based on type I collagen, and containing either strontium-enriched mesoporous bioactive glasses or rod-like hydroxyapatite nanoparticles. The developed nanostructured systems underwent genipin chemical crosslinking and were then tested with an indirect co-culture of human trabecular bone-derived OBs and buffy coat-derived OC precursors, for 2–3 weeks. The favorable structural and biological properties of the materials proved to successfully support the viability, adhesion, and differentiation of cells, encouraging a further investigation of the developed bioactive systems as biomaterial inks for the 3D printing of more complex scaffolds for BTE.


Author(s):  
Aimee G. Kakascik

Osteogenesis imperfecta (OI) is a genetic disorder that affects collagen formation and ultimately leads to increased bone fragility. The fragile nature of the bones leads to fractures, even from seemingly normal patient care. Affected patients are at the highest risk for unintentional fractures during perioperative care. There are several different types of OI. Type I is the most common. With the different types come varying degrees of severity. Types II and III are the more severe forms. The classic clinical triad seen in OI is blue sclerae, multiple fractures, and conductive hearing loss. The patient may have other systemic involvement beyond the fragile musculoskeletal system. It is imperative that the anesthesiologist be well-versed in the natural history and perioperative management of patients with OI in order to optimize care and minimize complications.


1992 ◽  
Vol 286 (1) ◽  
pp. 73-77 ◽  
Author(s):  
M Mörike ◽  
R E Brenner ◽  
G B Bushart ◽  
W M Teller ◽  
U Vetter

Collagen produced in vitro by bone cells isolated from 19 patients with different forms of osteogenesis imperfecta (OI) was analysed. Clinically, four patients were classified as OI type I, 10 patients as OI type III and five patients as OI type IV. Bone cells of 12 of the 19 OI patients produced structurally abnormal type I collagen. Electrophoretically uniformly slower migrating collagen type I alpha-chains were found in one case of OI type I, in seven cases of OI type III and in one case of OI type IV; two cultures of OI type III produced two different populations of collagen type I alpha-chains, and one culture of OI type IV showed reduction-sensitive dimer formation of alpha 1(I) chains, resulting from the inadequate incorporation of a cysteine residue into the triple helical domain of alpha 1(I). Quantitative analysis of collagen metabolism led to the distinction of two groups of cultured OI osteoblasts. In osteoblasts of OI type I, mainly production of collagen was decreased, whereas secretion, processing and pericellular accumulation of (pro)collagen type I was similar to that in control osteoblasts. In contrast, in osteoblasts of OI types III and IV, production as well as secretion, processing and pericellular accumulation of (pro)collagen type I were significantly decreased. Low levels of type I collagen were found irrespective of the presence or absence of structural abnormalities of collagen type I in all OI types.


2021 ◽  
Author(s):  
◽  
Victoria L. Gremminger

Osteogenesis imperfecta (OI), also commonly referred to as brittle bone disease, is a heritable connective tissue disorder occurring in roughly 1:15,000 births. OI arises as a result of mutations in the type I collagen genes, COL1A1 and COL1A2, approximately 85 [percent] of the time with the remaining 15 [percent] of cases arising from mutations in genes involved in posttranslational modification of type I collagen, osteoblast maturation or mineralization. OI is a heterogeneous disorder that can be classified into four major types with severity ranging from perinatal lethality to premature osteoporosis. As the name suggests, osteogenesis imperfecta, literally translating to imperfect bone formation, results in bone fragility with patients often experiencing many fractures throughout their lifetime. While bone fragility is the most prominent manifestation of OI, skeletal muscle weakness, cardiopulmonary complications, short stature, and craniofacial abnormalities are also common. There is currently no cure for OI and therapeutic options rely on mitigating symptoms, primarily through the use of bone anti-resorptive agents referred to as bisphosphonates. Although, current treatment options focus solely on bone health, skeletal muscle weakness is a common manifestation in OI, where 80 [percent] of patients with mild OI experience muscle force deficits, and with even higher percentages in patients with more clinically severe OI. Historically, OI muscle weakness was largely attributed to inactivity with recent studies highlighting its inherent nature in both patients and mouse models. Studies investigating the mechanisms by which skeletal muscle weakness arises in OI are limited, despite the large prevalence. My research sought to better understand OI muscle weakness primarily through the investigation of mitochondrial health in a mouse modeling a severe human type III OI (oim/oim), as mitochondria are important regulators of energy metabolism and overall cell health. We hypothesized that oim/oim mice, exhibiting severe skeletal muscle weakness would exhibit mitochondrial dysfunction suggesting a correlation between skeletal muscle and mitochondrial function. To test this hypothesis, we assessed mitochondrial function and content in the oim/oim mouse. One of our major findings was the observation that oim/oim mice exhibit [greater than] 50 [percent] reductions in gastrocnemius mitochondrial respiration rates relative to wildtype littermates. Additionally, we found that citrate synthase activity in oim/oim isolated gastrocnemius mitochondria was reduced relative to wildtype littermates. Furthermore, to determine if skeletal muscle mitochondrial function correlated with skeletal muscle severity, we evaluated mitochondrial respiration in a mouse model of mild OI (+/oim). We did not find differences between +/oim and WT gastrocnemius mitochondrial respiration suggesting that mitochondrial function does correlate with skeletal muscle function. Moreover, we did not observe changes in mitochondrial respiration in oim/oim liver and heart suggesting the mitochondrial dysfunction is not global in the oim/oim mouse. Additionally, we sought to investigate whole body metabolic alterations, as skeletal muscle comprises roughly 50 [percent] of body mass and is a significant contributor to the body's resting metabolic rate. We hypothesized that skeletal muscle mitochondrial dysfunction in the oim/oim mouse would lead to changes in metabolic parameters including altered substrate utilization, altered body composition, and changes in energy expenditure. Interestingly, we did not observe changes in substrate utilization, although we did note increased energy expenditure and subtle changes in body composition with oim/oim animals exhibiting reduced percentages of fat mass and increased percentages of lean mass relative to wildtype littermates. Overall, my research was the first to implicate mitochondrial dysfunction in the pathophysiology of OI using a mouse model of severe OI. This work has led to numerous studies in other mouse models evaluating mitochondrial function and energy metabolism. While there is more work to be done to further understand the mechanisms and correlation between mitochondrial dysfunction and skeletal muscle weakness in OI, this novel finding has initiated a new area of research in OI and has contributed to the overall understanding of OI muscle weakness.


Biomolecules ◽  
2021 ◽  
Vol 11 (10) ◽  
pp. 1493
Author(s):  
Malwina Botor ◽  
Agnieszka Fus-Kujawa ◽  
Marta Uroczynska ◽  
Karolina L. Stepien ◽  
Anna Galicka ◽  
...  

Osteogenesis Imperfecta (OI) is a group of connective tissue disorders with a broad range of phenotypes characterized primarily by bone fragility. The prevalence of OI ranges from about 1:15,000 to 1:20,000 births. Five types of the disease are commonly distinguished, ranging from a mild (type I) to a lethal one (type II). Types III and IV are severe forms allowing survival after the neonatal period, while type V is characterized by a mild to moderate phenotype with calcification of interosseous membranes. In most cases, there is a reduction in the production of normal type I collagen (col I) or the synthesis of abnormal collagen as a result of mutations in col I genes. Moreover, mutations in genes involved in col I synthesis and processing as well as in osteoblast differentiation have been reported. The currently available treatments try to prevent fractures, control symptoms and increase bone mass. Commonly used medications in OI treatment are bisphosphonates, Denosumab, synthetic parathyroid hormone and growth hormone for children therapy. The main disadvantages of these therapies are their relatively weak effectiveness, lack of effects in some patients or cytotoxic side effects. Experimental approaches, particularly those based on stem cell transplantation and genetic engineering, seem to be promising to improve the therapeutic effects of OI.


PLoS ONE ◽  
2021 ◽  
Vol 16 (8) ◽  
pp. e0255315
Author(s):  
Rachel Kohler ◽  
Carli A. Tastad ◽  
Amy Creecy ◽  
Joseph M. Wallace

Osteogenesis imperfecta (OI) is a hereditary bone disease where gene mutations affect Type I collagen formation resulting in osteopenia and increased fracture risk. There are several established mouse models of OI, but some are severe and result in spontaneous fractures or early animal death. The Amish Col1a2G610C/+ (G610C) mouse model is a newer, moderate OI model that is currently being used in a variety of intervention studies, with differing background strains, sexes, ages, and bone endpoints. This study is a comprehensive mechanical and architectural characterization of bone in G610C mice bred on a C57BL/6 inbred strain and will provide a baseline for future treatment studies. Male and female wild-type (WT) and G610C mice were euthanized at 10 and 16 weeks (n = 13–16). Harvested tibiae, femora, and L4 vertebrae were scanned via micro-computed tomography and analyzed for cortical and trabecular architectural properties. Femora and tibiae were then mechanically tested to failure. G610C mice had less bone but more highly mineralized cortical and trabecular tissue than their sex- and age-matched WT counterparts, with cortical cross-sectional area, thickness, and mineral density, and trabecular bone volume, mineral density, spacing, and number all differing significantly as a function of genotype (2 Way ANOVA with main effects of sex and genotype at each age). In addition, mechanical yield force, ultimate force, displacement, strain, and toughness were all significantly lower in G610C vs. WT, highlighting a brittle phenotype. This characterization demonstrates that despite being a moderate OI model, the Amish G610C mouse model maintains a distinctly brittle phenotype and is well-suited for use in future intervention studies.


Sign in / Sign up

Export Citation Format

Share Document