1
|
Infante A, Alcorta-Sevillano N, Macías I, Cabodevilla L, Medhat D, Lafaver B, Crawford TK, Phillips CL, Bueno AM, Sagastizabal B, Arroyo M, Campino A, Gerovska D, Araúzo-Bravo M, Gener B, Rodríguez CI. Galunisertib downregulates mutant type I collagen expression and promotes MSCs osteogenesis in pediatric osteogenesis imperfecta. Biomed Pharmacother 2024; 175:116725. [PMID: 38744219 DOI: 10.1016/j.biopha.2024.116725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Qualitative alterations in type I collagen due to pathogenic variants in the COL1A1 or COL1A2 genes, result in moderate and severe Osteogenesis Imperfecta (OI), a rare disease characterized by bone fragility. The TGF-β signaling pathway is overactive in OI patients and certain OI mouse models, and inhibition of TGF-β through anti-TGF-β monoclonal antibody therapy in phase I clinical trials in OI adults is rendering encouraging results. However, the impact of TGF-β inhibition on osteogenic differentiation of mesenchymal stem cells from OI patients (OI-MSCs) is unknown. The following study demonstrates that pediatric skeletal OI-MSCs have imbalanced osteogenesis favoring the osteogenic commitment. Galunisertib, a small molecule inhibitor (SMI) that targets the TGF-β receptor I (TβRI), favored the final osteogenic maturation of OI-MSCs. Mechanistically, galunisertib downregulated type I collagen expression in OI-MSCs, with greater impact on mutant type I collagen, and concomitantly, modulated the expression of unfolded protein response (UPR) and autophagy markers. In vivo, galunisertib improved trabecular bone parameters only in female oim/oim mice. These results further suggest that type I collagen is a tunable target within the bone ECM that deserves investigation and that the SMI, galunisertib, is a promising new candidate for the anti-TGF-β targeting for the treatment of OI.
Collapse
Affiliation(s)
- Arantza Infante
- Stem Cells and Advanced Therapies Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Natividad Alcorta-Sevillano
- Stem Cells and Advanced Therapies Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Iratxe Macías
- Stem Cells and Advanced Therapies Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Leire Cabodevilla
- Stem Cells and Advanced Therapies Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Dokki, Giza, Egypt
| | - Brittany Lafaver
- Department of Biochemistry, University of Missouri, Columbia, USA
| | - Tara K Crawford
- Department of Biochemistry, University of Missouri, Columbia, USA
| | | | - Ana M Bueno
- Department of Orthopedic Surgery, Getafe University Hospital, Madrid, Spain
| | | | - Maitane Arroyo
- Department of Traumatology, Basurto Hospital, Bilbao, Spain
| | - Ainara Campino
- Service of Pharmacy, Cruces University Hospital, Barakaldo, Spain
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine Research Group, Biogipuzkoa Health Research Institute, Donostia, Spain
| | - Marcos Araúzo-Bravo
- Computational Biology and Systems Biomedicine Research Group, Biogipuzkoa Health Research Institute, Donostia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48009, Spain; Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Spain
| | - Blanca Gener
- Stem Cells and Advanced Therapies Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Service of Genetics, Cruces University Hospital, Barakaldo, Spain
| | - Clara I Rodríguez
- Stem Cells and Advanced Therapies Group, Biobizkaia Health Research Institute, Barakaldo, Spain.
| |
Collapse
|
2
|
Yang YS, Sato T, Chaugule S, Ma H, Xie J, Gao G, Shim JH. AAV-based gene editing of type 1 collagen mutation to treat osteogenesis imperfecta. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102111. [PMID: 38261950 PMCID: PMC10797194 DOI: 10.1016/j.omtn.2023.102111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Osteogenesis imperfecta (OI) is a genetic disorder characterized by bone fragility, low bone mass, fractures, and extraskeletal manifestations. Since OI is commonly caused by single-nucleotide mutation(s) in the COL1A1 or COL1A2 genes encoding type I collagens, we developed a genome-editing strategy to correct a Col1a2 mutation in an OIM mouse model resembling a severe dominant form of human type III OI. Using a recombinant adeno-associated virus (rAAV), we delivered CRISPR-Cas9 to bone-forming osteoblast-lineage cells in the skeleton. Homology-directed repair (HDR)-mediated gene editing efficiency in these cells was improved when CRISPR-Cas9 was coupled with a donor AAV vector containing a promoterless partial mouse Col1a2 complementary DNA sequence. This approach effectively reversed the dysregulation of osteogenic differentiation by a Col1a2 mutation in vitro. Furthermore, systemic administration of dual rAAVs in OIM mice lowered bone matrix turnover rates by reducing osteoblast and osteoclast development while improving the cellular network of mechano-sensing osteocytes embedded in the bone matrix. This strategy significantly improved bone architecture/mass/mineralization, skeletal deformities, grip strength, and spontaneous fractures. Our study is the first demonstration that HDR-mediated gene editing via AAV-mediated delivery effectively corrects a collagen mutation in OI osteoblasts and reverses skeletal phenotypes in OIM mice.
Collapse
Affiliation(s)
- Yeon-Suk Yang
- Department of Medicine, Division of Rheumatology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Tadatoshi Sato
- Department of Medicine, Division of Rheumatology, UMass Chan Medical School, Worcester, MA 01655, USA
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01655, USA
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA 01655, USA
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sachin Chaugule
- Department of Medicine, Division of Rheumatology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Hong Ma
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01655, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Jun Xie
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01655, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01655, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01655, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA 01655, USA
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Jae-Hyuck Shim
- Department of Medicine, Division of Rheumatology, UMass Chan Medical School, Worcester, MA 01655, USA
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01655, USA
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
3
|
Dinulescu A, Păsărică AS, Carp M, Dușcă A, Dijmărescu I, Pavelescu ML, Păcurar D, Ulici A. New Perspectives of Therapies in Osteogenesis Imperfecta-A Literature Review. J Clin Med 2024; 13:1065. [PMID: 38398378 PMCID: PMC10888533 DOI: 10.3390/jcm13041065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Background: Osteogenesis imperfecta (OI) is a rare skeletal dysplasia characterized as a heterogeneous disorder group with well-defined phenotypic and genetic features that share uncommon bone fragility. The current treatment options, medical and orthopedic, are limited and not efficient enough to improve the low bone density, bone fragility, growth, and mobility of the affected individuals, creating the need for alternative therapeutic agents. (2) Methods: We searched the medical database to find papers regarding treatments for OI other than conventional ones. We included 45 publications. (3) Results: In reviewing the literature, eight new potential therapies for OI were identified, proving promising results in cells and animal models or in human practice, but further research is still needed. Bone marrow transplantation is a promising therapy in mice, adults, and children, decreasing the fracture rate with a beneficial effect on structural bone proprieties. Anti-RANKL antibodies generated controversial results related to the therapy schedule, from no change in the fracture rate to improvement in the bone mineral density resorption markers and bone formation, but with adverse effects related to hypercalcemia. Sclerostin inhibitors in murine models demonstrated an increase in the bone formation rate and trabecular cortical bone mass, and a few human studies showed an increase in biomarkers and BMD and the downregulation of resorption markers. Recombinant human parathormone and TGF-β generated good results in human studies by increasing BMD, depending on the type of OI. Gene therapy, 4-phenylbutiric acid, and inhibition of eIF2α phosphatase enzymes have only been studied in cell cultures and animal models, with promising results. (4) Conclusions: This paper focuses on eight potential therapies for OI, but there is not yet enough data for a new, generally accepted treatment. Most of them showed promising results, but further research is needed, especially in the pediatric field.
Collapse
Affiliation(s)
- Alexandru Dinulescu
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Alexandru-Sorin Păsărică
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Mădălina Carp
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Andrei Dușcă
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Irina Dijmărescu
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Mirela Luminița Pavelescu
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Daniela Păcurar
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| | - Alexandru Ulici
- Departament of Pediatrics and Department of Pediatric Orthopedics, “Carol Davila“ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.D.); (M.C.); (A.D.); (I.D.); (M.L.P.); (A.U.)
- Departament of Pediatrics and Department of Pediatric Orthopedics, Emergency Hospital for Children ‘’Grigore Alexandrescu’’, 011743 Bucharest, Romania;
| |
Collapse
|
4
|
Sun Y, Li L, Wang J, Liu H, Wang H. Emerging Landscape of Osteogenesis Imperfecta Pathogenesis and Therapeutic Approaches. ACS Pharmacol Transl Sci 2024; 7:72-96. [PMID: 38230285 PMCID: PMC10789133 DOI: 10.1021/acsptsci.3c00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/18/2024]
Abstract
Osteogenesis imperfecta (OI) is an uncommon genetic disorder characterized by shortness of stature, hearing loss, poor bone mass, recurrent fractures, and skeletal abnormalities. Pathogenic variations have been found in over 20 distinct genes that are involved in the pathophysiology of OI, contributing to the disorder's clinical and genetic variability. Although medications, surgical procedures, and other interventions can partially alleviate certain symptoms, there is still no known cure for OI. In this Review, we provide a comprehensive overview of genetic pathogenesis, existing treatment modalities, and new developments in biotechnologies such as gene editing, stem cell reprogramming, functional differentiation, and transplantation for potential future OI therapy.
Collapse
Affiliation(s)
- Yu Sun
- PET
Center, Chongqing University Three Gorges
Hospital, Chongqing 404000, China
| | - Lin Li
- PET
Center, Chongqing University Three Gorges
Hospital, Chongqing 404000, China
| | - Jiajun Wang
- Medical
School of Hubei Minzu University, Enshi 445000, China
| | - Huiting Liu
- PET
Center, Chongqing University Three Gorges
Hospital, Chongqing 404000, China
| | - Hu Wang
- Department
of Neurology, Johns Hopkins University School
of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
5
|
Lauffer MC, van Roon-Mom W, Aartsma-Rus A. Possibilities and limitations of antisense oligonucleotide therapies for the treatment of monogenic disorders. COMMUNICATIONS MEDICINE 2024; 4:6. [PMID: 38182878 PMCID: PMC10770028 DOI: 10.1038/s43856-023-00419-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are incredibly versatile molecules that can be designed to specifically target and modify RNA transcripts to slow down or halt rare genetic disease progression. They offer the potential to target groups of patients or can be tailored for individual cases. Nonetheless, not all genetic variants and disorders are amenable to ASO-based treatments, and hence, it is important to consider several factors before embarking on the drug development journey. Here, we discuss which genetic disorders have the potential to benefit from a specific type of ASO approach, based on the pathophysiology of the disease and pathogenic variant type, as well as those disorders that might not be suitable for ASO therapies. We further explore additional aspects, such as the target tissues, intervention time points, and potential clinical benefits, which need to be considered before developing a compound. Overall, we provide an overview of the current potentials and limitations of ASO-based therapeutics for the treatment of monogenic disorders.
Collapse
Affiliation(s)
- Marlen C Lauffer
- Dutch Center for RNA Therapeutics, Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Willeke van Roon-Mom
- Dutch Center for RNA Therapeutics, Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemieke Aartsma-Rus
- Dutch Center for RNA Therapeutics, Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
6
|
Schindeler A, Lee LR, O'Donohue AK, Ginn SL, Munns CF. Curative Cell and Gene Therapy for Osteogenesis Imperfecta. J Bone Miner Res 2022; 37:826-836. [PMID: 35306687 PMCID: PMC9324990 DOI: 10.1002/jbmr.4549] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/03/2022] [Accepted: 02/27/2022] [Indexed: 11/17/2022]
Abstract
Osteogenesis imperfecta (OI) describes a series of genetic bone fragility disorders that can have a substantive impact on patient quality of life. The multidisciplinary approach to management of children and adults with OI primarily involves the administration of antiresorptive medication, allied health (physiotherapy and occupational therapy), and orthopedic surgery. However, advances in gene editing technology and gene therapy vectors bring with them the promise of gene-targeted interventions to provide an enduring or perhaps permanent cure for OI. This review describes emergent technologies for cell- and gene-targeted therapies, major hurdles to their implementation, and the prospects of their future success with a focus on bone disorders. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Aaron Schindeler
- Bioengineering and Molecular Medicine Laboratorythe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadAustralia
- Children's Hospital Westmead Clinical SchoolUniversity of SydneyCamperdownAustralia
| | - Lucinda R Lee
- Bioengineering and Molecular Medicine Laboratorythe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadAustralia
- Children's Hospital Westmead Clinical SchoolUniversity of SydneyCamperdownAustralia
| | - Alexandra K O'Donohue
- Bioengineering and Molecular Medicine Laboratorythe Children's Hospital at Westmead and the Westmead Institute for Medical ResearchWestmeadAustralia
- Children's Hospital Westmead Clinical SchoolUniversity of SydneyCamperdownAustralia
| | - Samantha L Ginn
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and HealthThe University of Sydney and Sydney Children's Hospitals NetworkWestmeadAustralia
| | - Craig F Munns
- Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
- Department of Endocrinology and DiabetesQueensland Children's HospitalBrisbaneQLDAustralia
- Child Health Research Centre and Faculty of MedicineThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
7
|
Botor M, Fus-Kujawa A, Uroczynska M, Stepien KL, Galicka A, Gawron K, Sieron AL. Osteogenesis Imperfecta: Current and Prospective Therapies. Biomolecules 2021; 11:biom11101493. [PMID: 34680126 PMCID: PMC8533546 DOI: 10.3390/biom11101493] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Malwina Botor
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752 Katowice, Poland; (A.F.-K.); (M.U.); (K.L.S.); (K.G.); (A.L.S.)
- Correspondence:
| | - Agnieszka Fus-Kujawa
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752 Katowice, Poland; (A.F.-K.); (M.U.); (K.L.S.); (K.G.); (A.L.S.)
| | - Marta Uroczynska
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752 Katowice, Poland; (A.F.-K.); (M.U.); (K.L.S.); (K.G.); (A.L.S.)
| | - Karolina L. Stepien
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752 Katowice, Poland; (A.F.-K.); (M.U.); (K.L.S.); (K.G.); (A.L.S.)
| | - Anna Galicka
- Department of Medical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-222 Bialystok, Poland;
| | - Katarzyna Gawron
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752 Katowice, Poland; (A.F.-K.); (M.U.); (K.L.S.); (K.G.); (A.L.S.)
| | - Aleksander L. Sieron
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752 Katowice, Poland; (A.F.-K.); (M.U.); (K.L.S.); (K.G.); (A.L.S.)
| |
Collapse
|
8
|
Shen Z, Chen M, Luo F, Xu H, Zhang P, Lin J, Kang M. Identification of Key Genes and Pathways Associated With Paclitaxel Resistance in Esophageal Squamous Cell Carcinoma Based on Bioinformatics Analysis. Front Genet 2021; 12:671639. [PMID: 34456964 PMCID: PMC8386171 DOI: 10.3389/fgene.2021.671639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/02/2021] [Indexed: 01/07/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) ranks as the fourth leading cause of cancer-related death in China. Although paclitaxel has been shown to be effective in treating ESCC, the prolonged use of this chemical will lead to paclitaxel resistance. In order to uncover genes and pathways driving paclitaxel resistance in the progression of ESCC, bioinformatics analyses were performed based on The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database including GSE86099 and GSE161533. Differential expression analysis was performed in TCGA data and two GEO datasets to obtain differentially expressed genes (DEGs). Based on GSE161533, weighted gene co-expression network analysis (WGCNA) was conducted to identify the key modules associated with ESCC tumor status. The DEGs common to the two GEO datasets and the genes in the key modules were intersected to obtain the paclitaxel resistance-specific or non-paclitaxel resistance-specific genes, which were subjected to subsequent least absolute shrinkage and selection operator (LASSO) feature selection, whereby paclitaxel resistance-specific or non-paclitaxel resistance-specific key genes were selected. Ten machine learning models were used to validate the biological significance of these key genes; the potential therapeutic drugs for paclitaxel resistance-specific genes were also predicted. As a result, we identified 24 paclitaxel resistance-specific genes and 18 non-paclitaxel resistance-specific genes. The ESCC machine classifiers based on the key genes achieved a relatively high AUC value in the cross-validation and in an independent test set, GSE164158. A total of 207 drugs (such as bevacizumab) were predicted to be alternative therapeutics for ESCC patients with paclitaxel resistance. These results might shed light on the in-depth research of paclitaxel resistance in the context of ESCC progression.
Collapse
Affiliation(s)
- Zhimin Shen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingduan Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fei Luo
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hui Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jihong Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Trajanoska K, Rivadeneira F. Genomic Medicine: Lessons Learned From Monogenic and Complex Bone Disorders. Front Endocrinol (Lausanne) 2020; 11:556610. [PMID: 33162933 PMCID: PMC7581702 DOI: 10.3389/fendo.2020.556610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
Current genetic studies of monogenic and complex bone diseases have broadened our understanding of disease pathophysiology, highlighting the need for medical interventions and treatments tailored to the characteristics of patients. As genomic research progresses, novel insights into the molecular mechanisms are starting to provide support to clinical decision-making; now offering ample opportunities for disease screening, diagnosis, prognosis and treatment. Drug targets holding mechanisms with genetic support are more likely to be successful. Therefore, implementing genetic information to the drug development process and a molecular redefinition of skeletal disease can help overcoming current shortcomings in pharmaceutical research, including failed attempts and appalling costs. This review summarizes the achievements of genetic studies in the bone field and their application to clinical care, illustrating the imminent advent of the genomic medicine era.
Collapse
|
10
|
Maruelli S, Besio R, Rousseau J, Garibaldi N, Amiaud J, Brulin B, Layrolle P, Escriou V, Rossi A, Trichet V, Forlino A. Osteoblasts mineralization and collagen matrix are conserved upon specific Col1a2 silencing. Matrix Biol Plus 2020; 6-7:100028. [PMID: 33543025 PMCID: PMC7852305 DOI: 10.1016/j.mbplus.2020.100028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 11/10/2022] Open
Abstract
Classical osteogenesis imperfecta (OI) is an inherited rare brittle bone disease caused by dominant mutations in the COL1A1 or COL1A2 genes, encoding for the α chains of collagen type I. The definitive cure for the disease will require a gene therapy approach, aimed to correct or suppress the mutant allele. Interestingly, individuals lacking α2(I) chain and synthetizing collagen α1(I)3 homotrimers do not show bone phenotype, making appealing a bone specific COL1A2 silencing approach for OI therapy. To this aim, three different Col1a2-silencing RNAs (siRNAs), −3554, −3825 and −4125, selected at the 3′-end of the murine Col1a2 transcript were tested in vitro and in vivo. In murine embryonic fibroblasts Col1a2-siRNA-3554 was able to efficiently and specifically target the Col1a2 mRNA and to strongly reduce α2(I) chain expression. Its efficiency and specificity were also demonstrated in primary murine osteoblasts, whose mineralization was preserved. The efficiency of Col1a2-siRNA-3554 was proved also in vivo. Biphasic calcium phosphate implants loaded with murine mesenchymal stem cells were intramuscularly transplanted in nude mice and injected with Col1a2-siRNA-3554 three times a week for three weeks. Collagen α2 silencing was demonstrated both at mRNA and protein level and Masson's Trichrome staining confirmed the presence of newly formed collagen matrix. Our data pave the way for further investigation of Col1a2 silencing and siRNA delivery to the bone tissue as a possible strategy for OI therapy. Identification of a specific and efficient Col1a2 siRNA Silencing of Col1a2 allows osteoblasts mineralization. Col1a2 silencing is not impairing matrix deposition in vivo.
Collapse
Key Words
- BCP, biphasic calcium phosphate
- Collagen
- D-MEM, Dulbecco-modified Eagle's medium
- EDS, Ehlers Danlos syndrome
- EGFP, enhanced green fluorescent protein
- FBS, fetal bovine serum
- Gene therapy
- MEF, murine embryonic fibroblast
- MSC, mesenchymal stem cell
- NMD, nonsense mediated RNA decay
- OI, osteogenesis imperfecta
- Osteogenesis imperfecta
- PBS, phosphate buffered saline
- RNAi, RNA interference
- SDS, sodium dodecyl sulphate
- Silencing
- TRAP, tartrate-resistant acid phosphatase
- shRNA, short hairpin RNA
- siRNA
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Silvia Maruelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Julie Rousseau
- INSERM, Université de Nantes, UMR1238, Phy-Os, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Nadia Garibaldi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Jérôme Amiaud
- INSERM, Université de Nantes, UMR1238, Phy-Os, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Bénédicte Brulin
- INSERM, Université de Nantes, UMR1238, Phy-Os, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Pierre Layrolle
- INSERM, Université de Nantes, UMR1238, Phy-Os, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | | | - Antonio Rossi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Valerie Trichet
- INSERM, Université de Nantes, UMR1238, Phy-Os, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| |
Collapse
|
11
|
Wong MY, Shoulders MD. Targeting defective proteostasis in the collagenopathies. Curr Opin Chem Biol 2019; 50:80-88. [PMID: 31028939 DOI: 10.1016/j.cbpa.2019.02.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/18/2022]
Abstract
The collagenopathies are a diverse group of diseases caused primarily by mutations in collagen genes. The resulting disruptions in collagen biogenesis can impair development, cause cellular dysfunction, and severely impact connective tissues. Most existing treatment options only address patient symptoms. Yet, while the disease-causing genes and proteins themselves are difficult to target, increasing evidence suggests that resculpting the intracellular proteostasis network, meaning the machineries responsible for producing and ensuring the integrity of collagen, could provide substantial benefit. We present a proteostasis-focused perspective on the collagenopathies, emphasizing progress toward understanding how mechanisms of collagen proteostasis are disrupted in disease. In parallel, we highlight recent advances in small molecule approaches to tune endoplasmic reticulum proteostasis that may prove useful in these disorders.
Collapse
Affiliation(s)
- Madeline Y Wong
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States.
| |
Collapse
|
12
|
Lamandé SR, Bateman JF. Genetic Disorders of the Extracellular Matrix. Anat Rec (Hoboken) 2019; 303:1527-1542. [PMID: 30768852 PMCID: PMC7318566 DOI: 10.1002/ar.24086] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Mutations in the genes for extracellular matrix (ECM) components cause a wide range of genetic connective tissues disorders throughout the body. The elucidation of mutations and their correlation with pathology has been instrumental in understanding the roles of many ECM components. The pathological consequences of ECM protein mutations depend on its tissue distribution, tissue function, and on the nature of the mutation. The prevalent paradigm for the molecular pathology has been that there are two global mechanisms. First, mutations that reduce the production of ECM proteins impair matrix integrity largely due to quantitative ECM defects. Second, mutations altering protein structure may reduce protein secretion but also introduce dominant negative effects in ECM formation, structure and/or stability. Recent studies show that endoplasmic reticulum (ER) stress, caused by mutant misfolded ECM proteins, makes a significant contribution to the pathophysiology. This suggests that targeting ER‐stress may offer a new therapeutic strategy in a range of ECM disorders caused by protein misfolding mutations. Anat Rec, 2019. © 2019 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Shireen R Lamandé
- Musculoskeletal Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville Victoria, Australia
| | - John F Bateman
- Musculoskeletal Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville Victoria, Australia
| |
Collapse
|
13
|
Abstract
Steady-state and pulse-chase collagen analyses are powerful approaches to investigate in vitro the structure of collagen and its kinetic of secretion, respectively. The electrophoretic analysis of purified 3H-proline-labeled collagen allows to determine the nature and post translational modifications of its α chains, whereas short-pulse labeling can be used to follow collagen secretion over time.
Collapse
Affiliation(s)
- Antonella Forlino
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| | - Francesca Tonelli
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Roberta Besio
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
14
|
Morello R. Osteogenesis imperfecta and therapeutics. Matrix Biol 2018; 71-72:294-312. [PMID: 29540309 PMCID: PMC6133774 DOI: 10.1016/j.matbio.2018.03.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 02/08/2023]
Abstract
Osteogenesis imperfecta, or brittle bone disease, is a congenital disease that primarily causes low bone mass and bone fractures but it can negatively affect other organs. It is usually inherited in an autosomal dominant fashion, although rarer recessive and X-chromosome-linked forms of the disease have been identified. In addition to type I collagen, mutations in a number of other genes, often involved in type I collagen synthesis or in the differentiation and function of osteoblasts, have been identified in the last several years. Seldom, the study of a rare disease has delivered such a wealth of new information that have helped our understanding of multiple processes involved in collagen synthesis and bone formation. In this short review I will describe the clinical features and the molecular genetics of the disease, but then focus on how OI dysregulates all aspects of extracellular matrix biology. I will conclude with a discussion about OI therapeutics.
Collapse
Affiliation(s)
- Roy Morello
- Department of Physiology & Biophysics, Orthopaedic Surgery, and Division of Genetics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
15
|
Gioia R, Tonelli F, Ceppi I, Biggiogera M, Leikin S, Fisher S, Tenedini E, Yorgan TA, Schinke T, Tian K, Schwartz JM, Forte F, Wagener R, Villani S, Rossi A, Forlino A. The chaperone activity of 4PBA ameliorates the skeletal phenotype of Chihuahua, a zebrafish model for dominant osteogenesis imperfecta. Hum Mol Genet 2018; 26:2897-2911. [PMID: 28475764 PMCID: PMC5886106 DOI: 10.1093/hmg/ddx171] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022] Open
Abstract
Classical osteogenesis imperfecta (OI) is a bone disease caused by type I collagen mutations and characterized by bone fragility, frequent fractures in absence of trauma and growth deficiency. No definitive cure is available for OI and to develop novel drug therapies, taking advantage of a repositioning strategy, the small teleost zebrafish (Danio rerio) is a particularly appealing model. Its small size, high proliferative rate, embryo transparency and small amount of drug required make zebrafish the model of choice for drug screening studies, when a valid disease model is available. We performed a deep characterization of the zebrafish mutant Chihuahua, that carries a G574D (p.G736D) substitution in the α1 chain of type I collagen. We successfully validated it as a model for classical OI. Growth of mutants was delayed compared with WT. X-ray, µCT, alizarin red/alcian blue and calcein staining revealed severe skeletal deformity, presence of fractures and delayed mineralization. Type I collagen extracted from different tissues showed abnormal electrophoretic migration and low melting temperature. The presence of endoplasmic reticulum (ER) enlargement due to mutant collagen retention in osteoblasts and fibroblasts of mutant fish was shown by electron and confocal microscopy. Two chemical chaperones, 4PBA and TUDCA, were used to ameliorate the cellular stress and indeed 4PBA ameliorated bone mineralization in larvae and skeletal deformities in adult, mainly acting on reducing ER cisternae size and favoring collagen secretion. In conclusion, our data demonstrated that ER stress is a novel target to ameliorate OI phenotype; chemical chaperones such as 4PBA may be, alone or in combination, a new class of molecules to be further investigated for OI treatment.
Collapse
Affiliation(s)
- Roberta Gioia
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Francesca Tonelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Ilaria Ceppi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Marco Biggiogera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Sergey Leikin
- Section on Physical Biochemistry, Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD, USA
| | - Shannon Fisher
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Elena Tenedini
- Center for Genome Research, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Timur A Yorgan
- Institute of Osteology and Biomechanic, Center for Experimental Medicine, University of Hamburg, Hamburg, Germany
| | - Thorsten Schinke
- Institute of Osteology and Biomechanic, Center for Experimental Medicine, University of Hamburg, Hamburg, Germany
| | - Kun Tian
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jean-Marc Schwartz
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Fabiana Forte
- Medical Faculty, Center for Biochemistry, Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Raimund Wagener
- Medical Faculty, Center for Biochemistry, Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Simona Villani
- Department of Public Health and Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Antonio Rossi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| |
Collapse
|
16
|
Marom R, Lee YC, Grafe I, Lee B. Pharmacological and biological therapeutic strategies for osteogenesis imperfecta. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2016; 172:367-383. [PMID: 27813341 DOI: 10.1002/ajmg.c.31532] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Osteogenesis imperfecta (OI) is a connective tissue disorder characterized by bone fragility, low bone mass, and bone deformities. The majority of cases are caused by autosomal dominant pathogenic variants in the COL1A1 and COL1A2 genes that encode type I collagen, the major component of the bone matrix. The remaining cases are caused by autosomal recessively or dominantly inherited mutations in genes that are involved in the post-translational modification of type I collagen, act as type I collagen chaperones, or are members of the signaling pathways that regulate bone homeostasis. The main goals of treatment in OI are to decrease fracture incidence, relieve bone pain, and promote mobility and growth. This requires a multi-disciplinary approach, utilizing pharmacological interventions, physical therapy, orthopedic surgery, and monitoring nutrition with appropriate calcium and vitamin D supplementation. Bisphosphonate therapy, which has become the mainstay of treatment in OI, has proven beneficial in increasing bone mass, and to some extent reducing fracture risk. However, the response to treatment is not as robust as is seen in osteoporosis, and it seems less effective in certain types of OI, and in adult OI patients as compared to most pediatric cases. New pharmacological treatments are currently being developed, including anti-resorptive agents, anabolic treatment, and gene- and cell-therapy approaches. These therapies are under different stages of investigation from the bench-side, to pre-clinical and clinical trials. In this review, we will summarize the recent findings regarding the pharmacological and biological strategies for the treatment of patients with OI. © 2016 Wiley Periodicals, Inc.
Collapse
|
17
|
Bishop N. Bone Material Properties in Osteogenesis Imperfecta. J Bone Miner Res 2016; 31:699-708. [PMID: 26987995 DOI: 10.1002/jbmr.2835] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/08/2016] [Accepted: 03/10/2016] [Indexed: 12/29/2022]
Abstract
Osteogenesis imperfecta entrains changes at every level in bone tissue, from the disorganization of the collagen molecules and mineral platelets within and between collagen fibrils to the macroarchitecture of the whole skeleton. Investigations using an array of sophisticated instruments at multiple scale levels have now determined many aspects of the effect of the disease on the material properties of bone tissue. The brittle nature of bone in osteogenesis imperfecta reflects both increased bone mineralization density-the quantity of mineral in relation to the quantity of matrix within a specific bone volume-and altered matrix-matrix and matrix mineral interactions. Contributions to fracture resistance at multiple scale lengths are discussed, comparing normal and brittle bone. Integrating the available information provides both a better understanding of the effect of current approaches to treatment-largely improved architecture and possibly some macroscale toughening-and indicates potential opportunities for alternative strategies that can influence fracture resistance at longer-length scales.
Collapse
Affiliation(s)
- Nick Bishop
- University of Sheffield and Sheffield Children's NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
18
|
Besio R, Forlino A. New frontiers for dominant osteogenesis imperfecta treatment: gene/cellular therapy approaches. ACTA ACUST UNITED AC 2015. [DOI: 10.3402/arb.v2.27964] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
|