1
|
Abstract
Viral gene transfer, known as transduction, is a powerful research tool for studying the biology of chondrocytes in novel ways and also a technology enabling the use of gene therapy for regenerating cartilage and treating diseases that affect cartilage, such as osteoarthritis. Adenovirus, retrovirus, lentivirus, and adeno-associated virus (AAV) are most commonly used to transduce chondrocytes. Although AAV is able to transduce chondrocytes in situ by intra-articular injection, chondrocytes are most commonly transduced in monolayer culture using the four vectors mentioned above. Protocols for achieving this are described, along with a discussion of the variables that can influence transduction efficiency.
Collapse
Affiliation(s)
| | - Christopher H Evans
- Musculoskeletal Gene Therapy Laboratory, Mayo Clinic, Rochester, MN, USA.
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN, USA.
| | | |
Collapse
|
2
|
Kondratova L, Kondratov O, Ragheb R, Zolotukhin S. Removal of Endotoxin from rAAV Samples Using a Simple Detergent-Based Protocol. Mol Ther Methods Clin Dev 2019; 15:112-119. [PMID: 31649960 PMCID: PMC6804492 DOI: 10.1016/j.omtm.2019.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/28/2019] [Indexed: 12/19/2022]
Abstract
Endotoxin is the most common contaminant found in protein samples. Even a small amount of endotoxin can induce strong allergic reaction and death of a host organism. Endotoxin is also often detected in recombinant adeno-associated virus (rAAV) stocks prepared in research laboratories using off-the-shelf reagents; purifying rAAV stocks from endotoxin using commercial reagents sometimes results in significant titer loss. The problem is exacerbated due to the recently expanded diversity of rAAV serotypes and capsid variants, which, due to their variable capsid surface charge, display differential affinity toward endotoxin. In this paper, we describe a simple universal protocol of purifying vector stocks irrespective of AAV serotype. The protocol is based on subjecting endotoxin-contaminated rAAV to mild detergent treatment, followed by repeated buffer-exchange washing and concentrating viral stock by low-speed centrifugation. Multiple assays were employed to test the physical and biological equivalency of the viral stocks before and after purification. The described protocol has been routinely utilized to purify vector stocks contaminated at levels as high as >1,000 endotoxin units (EU)/mL to produce viral vectors with practically undetectable levels of endotoxin (<2.5 EU/mL), with the titer's recovery in the range of 50%-100%.
Collapse
Affiliation(s)
- Liudmyla Kondratova
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville 32610, FL, USA
| | - Oleksandr Kondratov
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville 32610, FL, USA
| | | | - Sergei Zolotukhin
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville 32610, FL, USA
| |
Collapse
|
3
|
Tahaei SE, Couasnay G, Ma Y, Paria N, Gu J, Lemoine BF, Wang X, Rios JJ, Elefteriou F. The reduced osteogenic potential of Nf1-deficient osteoprogenitors is EGFR-independent. Bone 2018; 106:103-111. [PMID: 29032173 PMCID: PMC5694354 DOI: 10.1016/j.bone.2017.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/28/2017] [Accepted: 10/10/2017] [Indexed: 12/26/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common genetic disorder caused by mutations in the NF1 gene. Recalcitrant bone healing following fracture (i.e. pseudarthrosis) is one of the most problematic skeletal complications associated with NF1. The etiology of this condition is still unclear; thus, pharmacological options for clinical management are limited. Multiple studies have shown the reduced osteogenic potential of Nf1-deficient osteoprogenitors. A recent transcriptome profiling investigation revealed that EREG and EGFR, encoding epiregulin and its receptor Epidermal Growth Factor Receptor 1, respectively, were among the top over-expressed genes in cells of the NF1 pseudarthrosis site. Because EGFR stimulation is known to inhibit osteogenic differentiation, we hypothesized that increased EREG and EGFR expression in NF1-deficient skeletal progenitors may contribute to their reduced osteogenic differentiation potential. In this study, we first confirmed via single-cell mRNA sequencing that EREG over-expression was associated with NF1 second hit somatic mutations in human bone cells, whereas Transforming Growth Factor beta 1 (TGFβ1) expression was unchanged. Second, using ex-vivo recombined Nf1-deficient mouse bone marrow stromal cells (mBMSCs), we show that this molecular signature is conserved between mice and humans, and that epiregulin generated by these cells is overexpressed and active, whereas soluble TGFβ1 expression and activity are not affected. However, blocking either epiregulin function or EGFR signaling by EGFR1 or pan EGFR inhibition (using AG-1478 and Poziotinib respectively) did not correct the differentiation defect of Nf1-deficient mBMSCs, as measured by the expression of Alpl, Ibsp and alkaline phosphatase activity. These results suggest that clinically available drugs aimed at inhibiting EGFR signaling are unlikely to have a significant benefit for the management of bone non-union in children with NF1 PA.
Collapse
Affiliation(s)
- S E Tahaei
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - G Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Y Ma
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - N Paria
- Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, United States
| | - J Gu
- Baylor Institute for Immunology Research, Dallas, TX, United States
| | - B F Lemoine
- Baylor Institute for Immunology Research, Dallas, TX, United States
| | - X Wang
- Baylor Institute for Immunology Research, Dallas, TX, United States
| | - J J Rios
- Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, United States; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States; McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, United States; Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - F Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
4
|
Alvarez-Urena P, Zhu B, Henslee G, Sonnet C, Davis E, Sevick-Muraca E, Davis A, Olmsted-Davis E. Development of a Cell-Based Gene Therapy Approach to Selectively Turn Off Bone Formation. J Cell Biochem 2017. [PMID: 28621436 DOI: 10.1002/jcb.26220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cell and gene therapy approaches are safer when they possess a system that enables the therapy to be rapidly halted. Human mesenchymal stem cells were transduced with an adenoviral vector containing the cDNA for bone morphogenetic protein 2 (AdBMP2) to induce bone formation. To make this method safer, a system to quickly kill these virally transduced cells was designed and evaluated. Cells were encapsulated inside poly(ethylene glycol) diacrylate (PEG-Da) hydrogels that are able to shield the cells from immunological destruction. The system involves an inducible caspase-9 (iCasp9) activated using a specific chemical inducer of dimerization (CID). Delivering AdBMP2-transduced human mesenchymal stem cells encapsulated in PEG-Da hydrogel promoted ectopic ossification in vivo, and the iCasp9 system allowed direct control of the timing of apoptosis of the injected cells. The iCasp9-CID system enhances the safety of delivering AdBMP2-transduced cells, making it a more compelling therapeutic for bone repair and spine fusion. J. Cell. Biochem. 118: 3627-3634, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pedro Alvarez-Urena
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Banghe Zhu
- Center for Molecular Imaging, University of Texas Health Sciences Center, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas
| | - Gabrielle Henslee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Corinne Sonnet
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eleanor Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eva Sevick-Muraca
- Center for Molecular Imaging, University of Texas Health Sciences Center, Houston, Texas
| | - Alan Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| | - Elizabeth Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
5
|
Olmsted-Davis EA, Salisbury EA, Hoang D, Davis EL, Lazard Z, Sonnet C, Davis TA, Forsberg JA, Davis AR. Progenitors in Peripheral Nerves Launch Heterotopic Ossification. Stem Cells Transl Med 2017; 6:1109-1119. [PMID: 28198109 PMCID: PMC5442844 DOI: 10.1002/sctm.16-0347] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/31/2016] [Indexed: 12/23/2022] Open
Abstract
Studies presented here, using a murine model of bone morphogenetic protein type 2 (BMP2)-induced heterotopic ossification (HO) show that the protein initiates HO by signaling through progenitors in the endoneurium of peripheral nerves. In the mouse, these cells were identified in the endoneurium one day after BMP2 induction using antibody against phosphoSMAD (PS) 1, 5, and 8. Studies conducted in a tracking mouse that contains a tamoxifen-regulated Wnt1-Cre recombinase crossed with a td Tomato red (TR) reporter (Wnt1CreErt :Ai9Tm) confirmed their neural origin. In this model both BMP2 induction and tamoxifen are absolutely required to induce TR. SP7+ (osterix+ )TR+ cells were found in the endoneurium on day 1 and associated with bone on day 7. Quantification of TR+ and TR- cells isolated by fluorescence-activated cell sorting showed that all SP7+ cells were found in the TR+ population, whereas only about 80% of the TR+ cells expressed SP7. Pre-chondrocytes (Sox 9+ ) and transient brown fat (tBAT, UCP1+ ) also coexpressed TR, suggesting that the progenitor in nerves is multi-potential. The endoneurium of human nerves near the site of HO contained many PS+ cells, and SP7+ cells were found in nerves and on bone in tissue from patients with HO. Control tissues and nerves did not contain these PS+ and SP7+ cells. Some osteoblasts on bone from patients with HO were positive for PS, suggesting the continued presence of BMP during bone formation. The data suggests that the progenitors for HO are derived from the endoneurium in both the mouse model of HO and in humans with HO. Stem Cells Translational Medicine 2017;6:1109-1119.
Collapse
Affiliation(s)
- Elizabeth A Olmsted-Davis
- Center for Cell and Gene Therapy.,Departments of Pediatrics and Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | - Thomas A Davis
- Department of Surgery, Uniformed Services University of the Health Sciences & the Walter Reed National Military Medical Center, Bethesda, Maryland, USA.,Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Jonathan A Forsberg
- Department of Surgery, Uniformed Services University of the Health Sciences & the Walter Reed National Military Medical Center, Bethesda, Maryland, USA.,Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Alan R Davis
- Center for Cell and Gene Therapy.,Departments of Pediatrics and Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Alvarez-Urena P, Davis E, Sonnet C, Henslee G, Gugala Z, Strecker EV, Linscheid LJ, Cuchiara M, West J, Davis A, Olmsted-Davis E. Encapsulation of Adenovirus BMP2-Transduced Cells with PEGDA Hydrogels Allows Bone Formation in the Presence of Immune Response. Tissue Eng Part A 2017; 23:177-184. [PMID: 27967655 DOI: 10.1089/ten.tea.2016.0277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Gene therapy approaches have been difficult to implement due to pre-existing immunity against the virus used for delivery. To circumvent this problem, a cell-based approach was developed that avoided the use of free virus within the animal. However, even cells transduced in vitro with E1- to E3-deleted adenovirus encoding bone morphogenetic protein 2 (AdBMP2) resulted in the production of virus-neutralizing antibodies in mice. Furthermore, when mice received an intramuscular injection of nonencoding adenovirus (AdEmpty)-transduced cells, AdBMP2-transduced cells were unable to launch bone formation when an intramuscular injection of these BMP2-producing cells was delivered 1 week later. This phenomenon was not observed in NOD/SCID mice, and could be overcome in C57BL/6 mice by encapsulating the adenovirus-transduced cells in a nondegradable hydrogel poly(ethylene glycol) diacrylate (PEGDA). Data collectively suggest that PEGDA hydrogel encapsulation of AdBMP2-transduced cells prevents pre-existing immunity from suppressing BMP2-induced bone formation.
Collapse
Affiliation(s)
- Pedro Alvarez-Urena
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Eleanor Davis
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Corinne Sonnet
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Gabrielle Henslee
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas
| | - Zbigniew Gugala
- 2 Department of Orthopedic Surgery and Rehabilitation, The University of Texas Medical Branch at Galveston , Galveston, Texas
| | - Edward V Strecker
- 2 Department of Orthopedic Surgery and Rehabilitation, The University of Texas Medical Branch at Galveston , Galveston, Texas
| | - Laura J Linscheid
- 2 Department of Orthopedic Surgery and Rehabilitation, The University of Texas Medical Branch at Galveston , Galveston, Texas
| | - Maude Cuchiara
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Jennifer West
- 3 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Alan Davis
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,4 Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine , Houston, Texas.,5 Department of Orthopedic Surgery, Baylor College of Medicine , Houston, Texas
| | - Elizabeth Olmsted-Davis
- 1 Center for Cell and Gene Therapy , Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas.,4 Department of Pediatrics-Section Hematology/Oncology, Baylor College of Medicine , Houston, Texas.,5 Department of Orthopedic Surgery, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
7
|
Davis EL, Sonnet C, Lazard ZW, Henslee G, Gugala Z, Salisbury EA, Strecker EV, Davis TA, Forsberg JA, Davis AR, Olmsted‐Davis EA. Location-dependent heterotopic ossification in the rat model: The role of activated matrix metalloproteinase 9. J Orthop Res 2016; 34:1894-1904. [PMID: 26919547 PMCID: PMC5001934 DOI: 10.1002/jor.23216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/23/2016] [Indexed: 02/04/2023]
Abstract
Extremity amputation or traumatic injury can often lead to the formation of heterotopic ossification (HO). Studies to induce HO in rat muscle using cell-based gene therapy show that this process appears to be location dependent. In the present study, HO was induced in mice and rats through injection of immunologically matched cells transduced with either a replication-defective adenovirus possessing bone morphogenetic protein 2 (BMP2) or an empty adenovirus vector (control). Injection in rat near the skeletal bone resulted in HO, whereas cells injected into the same muscle group but distal from the bone did not result in bone formation. When cells were injected in the same limb at both locations at the same time, HO was formed at both sites. Characterization of the bone formation in rats versus mice demonstrated that different sources of osteogenic progenitors were involved, which may account for the location dependent bone formation observed in the rat. Further experimentation has shown that a potential reason for this difference may be the inability of rat to activate matrix metalloproteinase 9 (MMP9), an essential protease in mice necessary for recruitment of progenitors. Inhibition of active MMP9 in mice led to a significant decrease in HO. The studies reported here provide insight into the mechanisms and pathways leading to bone formation in different animals and species. It appears that not all animal models are appropriate for testing HO therapies, and our studies also challenge the conventional wisdom that larger animal models are better for testing treatments affecting bone. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1894-1904, 2016.
Collapse
Affiliation(s)
- Eleanor L. Davis
- Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexas77030
| | - Corinne Sonnet
- Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexas77030,Department of MedicineBaylor College of MedicineHoustonTexas77030
| | | | - Gabrielle Henslee
- Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexas77030
| | - Zbigniew Gugala
- Department of Orthopedic SurgeryUniversity of Texas Medical Branch GalvestonGalvestonTexas77555
| | - Elizabeth A. Salisbury
- Department of Orthopedic SurgeryUniversity of Texas Medical Branch GalvestonGalvestonTexas77555
| | - Edward V. Strecker
- Department of Orthopedic SurgeryUniversity of Texas Medical Branch GalvestonGalvestonTexas77555
| | - Thomas A. Davis
- Department of Regenerative MedicineNaval Medical Research CenterSilver SpringMaryland20910
| | - Jonathan A. Forsberg
- Department of Regenerative MedicineNaval Medical Research CenterSilver SpringMaryland20910
| | - Alan R. Davis
- Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexas77030,Department of PediatricsBaylor College of MedicineHoustonTexas77030,Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas77030
| | - Elizabeth A. Olmsted‐Davis
- Center for Cell and Gene TherapyBaylor College of MedicineHoustonTexas77030,Department of PediatricsBaylor College of MedicineHoustonTexas77030,Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas77030
| |
Collapse
|
8
|
Davis EL, Salisbury EA, Olmsted-Davis E, Davis AR. Anaplerotic Accumulation of Tricarboxylic Acid Cycle Intermediates as Well as Changes in Other Key Metabolites During Heterotopic Ossification. J Cell Biochem 2015; 117:1044-53. [PMID: 26627193 PMCID: PMC4784167 DOI: 10.1002/jcb.25454] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/01/2015] [Indexed: 12/23/2022]
Abstract
Heterotopic ossification (HO) is the de novo formation of bone that occurs in soft tissue, through recruitment, expansion, and differentiation of multiple cells types including transient brown adipocytes, osteoblasts, chondrocytes, mast cells, and platelets to name a few. Much evidence is accumulating that suggests changes in metabolism may be required to accomplish this bone formation. Recent work using a mouse model of heterotopic bone formation reliant on delivery of adenovirus‐transduced cells expressing low levels of BMP2 showed the immediate expansion of a unique brown adipocyte‐like cell. These cells are undergoing robust uncoupled oxidative phosphorylation to a level such that oxygen in the microenvironment is dramatically lowered creating areas of hypoxia. It is unclear how these oxygen changes ultimately affect metabolism and bone formation. To identify the processes and changes occurring over the course of bone formation, HO was established in the mice, and tissues isolated at early and late times were subjected to a global metabolomic screen. Results show that there are significant changes in both glucose levels, as well as TCA cycle intermediates. Additionally, metabolites necessary for oxidation of stored lipids were also found to be significantly elevated. The complete results of this screen are presented here, and provide a unique picture of the metabolic changes occurring during heterotopic bone formation. J. Cell. Biochem. 117: 1044–1053, 2016. © 2015 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eleanor L Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, 77030
| | | | - Elizabeth Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, 77030.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, 77030
| | - Alan R Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, 77030.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030.,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, 77030
| |
Collapse
|
9
|
El-Hoss J, Cheng T, Carpenter EC, Sullivan K, Deo N, Mikulec K, Little DG, Schindeler A. A Combination of rhBMP-2 (Recombinant Human Bone Morphogenetic Protein-2) and MEK (MAP Kinase/ERK Kinase) Inhibitor PD0325901 Increases Bone Formation in a Murine Model of Neurofibromatosis Type I Pseudarthrosis. J Bone Joint Surg Am 2014; 96:e117. [PMID: 25031379 DOI: 10.2106/jbjs.m.00862] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Congenital tibial dysplasia is a severe pediatric condition that classically results in a persistent pseudarthrosis. A majority of these cases are associated with neurofibromatosis type I (NF1), a genetic disorder in which inactivation of the NF1 gene leads to overactivity of the Ras-MEK-MAPK (mitogen-activated protein kinase) signaling pathway. We therefore hypothesized that pharmaceutical inhibition of MEK-MAPK may be a beneficial therapeutic strategy. METHODS In vitro methods were used to demonstrate a role for the MEK inhibitor PD0325901 in promoting osteogenic differentiation in Nf1-/- calvarial osteoblasts. Local applications of rhBMP-2 and/or PD0325901 were then tested in a mouse model of NF1 tibial pseudarthrosis featuring localized double inactivation of the Nf1 gene in a fracture. Mice received no treatment, PD0325901 (10 mg/kg/day from two days before fracture to ten days after fracture), rhBMP-2 (10 μg), or a combination of rhBMP-2 and PD0325901. RESULTS Animals treated with the delivery vehicle alone, PD0325901, rhBMP-2, or the PD0325901 + rhBMP-2 combination showed union rates of 0%, 8%, 69% (p < 0.01), or 80% (p < 0.01), respectively, at twenty-one days after fracture. Mice treated with the rhBMP-2 + PD0325901 combination displayed a callus volume sixfold greater than the vehicle controls and twofold greater than the group receiving rhBMP-2 alone. Although MEK inhibition combined with rhBMP-2 led to increases in bone formation and union, the proportion of fibrous tissue in the callus was not significantly reduced. CONCLUSIONS The data suggest that MEK inhibition can promote bone formation in combination with rhBMP-2 in the context of an NF1 pseudarthrosis. However, PD0325901 did not promote substantive bone anabolism in the absence of an exogenous anabolic stimulus and did not suppress fibrosis. CLINICAL RELEVANCE This study examines a signaling pathway-based approach to treating poor bone healing in a model of NF1 pseudarthrosis.
Collapse
Affiliation(s)
- J El-Hoss
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - T Cheng
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - E C Carpenter
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - K Sullivan
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - N Deo
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - K Mikulec
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - D G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - A Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| |
Collapse
|
10
|
Neben CL, Idoni B, Salva JE, Tuzon CT, Rice JC, Krakow D, Merrill AE. Bent bone dysplasia syndrome reveals nucleolar activity for FGFR2 in ribosomal DNA transcription. Hum Mol Genet 2014; 23:5659-71. [PMID: 24908667 DOI: 10.1093/hmg/ddu282] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor receptor 2 (FGFR2) promotes osteoprogenitor proliferation and differentiation during bone development, yet how the receptor elicits these distinct cellular responses remains unclear. Analysis of the FGFR2-skeletal disorder bent bone dysplasia syndrome (BBDS) demonstrates that FGFR2, in addition to its canonical signaling activities at the plasma membrane, regulates bone formation from within the nucleolus. Previously, we showed that the unique FGFR2 mutations that cause BBDS reduce receptor levels at the plasma membrane and diminish responsiveness to extracellular FGF2. In this study, we find that these mutations, despite reducing canonical signaling, enhance nucleolar occupancy of FGFR2 at the ribosomal DNA (rDNA) promoter. Nucleolar FGFR2 activates rDNA transcription via interactions with FGF2 and UBF1 by de-repressing RUNX2. An increase in the nucleolar activity of FGFR2 in BBDS elevates levels of ribosomal RNA in the developing bone, consequently promoting osteoprogenitor cell proliferation and decreasing differentiation. Identifying FGFR2 as a transcriptional regulator of rDNA in bone unexpectedly reveals a nucleolar route for FGF signaling that allows for independent regulation of osteoprogenitor cell proliferation and differentiation.
Collapse
Affiliation(s)
- Cynthia L Neben
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Idoni
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and
| | - Joanna E Salva
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Creighton T Tuzon
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Judd C Rice
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Deborah Krakow
- Departments of Orthopaedic Surgery, Human Genetics, Pediatrics and Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA,
| |
Collapse
|
11
|
Evaluation of optimal concentration and exposure duration of valproic acid alone or in combination with ViraDuctin to augment adenovirus transduction in human adipose stem cells. Cytotherapy 2014; 16:612-8. [DOI: 10.1016/j.jcyt.2013.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 08/10/2013] [Accepted: 08/14/2013] [Indexed: 11/30/2022]
|
12
|
Sonnet C, Simpson CL, Olabisi RM, Sullivan K, Lazard Z, Gugala Z, Peroni JF, Weh JM, Davis AR, West JL, Olmsted-Davis EA. Rapid healing of femoral defects in rats with low dose sustained BMP2 expression from PEGDA hydrogel microspheres. J Orthop Res 2013; 31:1597-604. [PMID: 23832813 DOI: 10.1002/jor.22407] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 05/13/2013] [Indexed: 02/04/2023]
Abstract
Current strategies for bone regeneration after traumatic injury often fail to provide adequate healing and integration. Here, we combined the poly (ethylene glycol) diacrylate (PEGDA) hydrogel with allogeneic "carrier" cells transduced with an adenovirus expressing BMP2. The system is unique in that the biomaterial encapsulates the cells, shielding them and thus suppressing destructive inflammatory processes. Using this system, complete healing of a 5 mm-long femur defect in a rat model occurs in under 3 weeks, through secretion of 100-fold lower levels of protein as compared to doses of recombinant BMP2 protein used in studies which lead to healing in 2-3 months. New bone formation was evaluated radiographically, histologically, and biomechanically at 2, 3, 6, 9, and 12 weeks after surgery. Rapid bone formation bridged the defect area and reliably integrated into the adjacent skeletal bone as early as 2 weeks. At 3 weeks, biomechanical analysis showed the new bone to possess 79% of torsional strength of the intact contralateral femur. Histological evaluation showed normal bone healing, with no infiltration of inflammatory cells with the bone being stable approximately 1 year later. We propose that these osteoinductive microspheres offer a more efficacious and safer clinical option over the use of rhBMP2.
Collapse
Affiliation(s)
- Corinne Sonnet
- Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Alkek Building, Room N1010, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Salisbury EA, Lazard ZW, Ubogu EE, Davis AR, Olmsted-Davis EA. Transient brown adipocyte-like cells derive from peripheral nerve progenitors in response to bone morphogenetic protein 2. Stem Cells Transl Med 2012; 1:874-85. [PMID: 23283549 DOI: 10.5966/sctm.2012-0090] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Perineurial-associated brown adipocyte-like cells were rapidly generated during bone morphogenetic protein 2 (BMP2)-induced sciatic nerve remodeling in the mouse. Two days after intramuscular injection of transduced mouse fibroblast cells expressing BMP2 into wild-type mice, there was replication of beta-3 adrenergic receptor(+) (ADRB3(+)) cells within the sciatic nerve perineurium. Fluorescence-activated cell sorting and analysis of cells isolated from these nerves confirmed ADRB3(+) cell expansion and their expression of the neural migration marker HNK1. Similar analysis performed 4 days after BMP2 delivery revealed a significant decrease in ADRB3(+) cells from isolated sciatic nerves, with their concurrent appearance within the adjacent soft tissue, suggesting migration away from the nerve. These soft tissue-derived cells also expressed the brown adipose marker uncoupling protein 1 (UCP1). Quantification of ADRB3-specific RNA in total hind limb tissue revealed a 3-fold increase 2 days after delivery of BMP2, followed by a 70-fold increase in UCP1-specific RNA after 3 days. Expression levels then rapidly returned to baseline by 4 days. Interestingly, these ADRB3(+) UCP1(+) cells also expressed the neural guidance factor reelin. Reelin(+) cells demonstrated distinct patterns within the injected muscle, concentrated toward the area of BMP2 release. Blocking mast cell degranulation-induced nerve remodeling resulted in the complete abrogation of UCP1-specific RNA and protein expression within the hind limbs following BMP2 injection. The data collectively suggest that local BMP2 administration initiates a cascade of events leading to the expansion, migration, and differentiation of progenitors from the peripheral nerve perineurium to brown adipose-like cells in the mouse, a necessary prerequisite for associated nerve remodeling.
Collapse
|
14
|
Salisbury E, Rodenberg E, Sonnet C, Hipp J, Gannon FH, Vadakkan TJ, Dickinson ME, Olmsted-Davis EA, Davis AR. Sensory nerve induced inflammation contributes to heterotopic ossification. J Cell Biochem 2012; 112:2748-58. [PMID: 21678472 DOI: 10.1002/jcb.23225] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Heterotopic ossification (HO), or bone formation in soft tissues, is often the result of traumatic injury. Much evidence has linked the release of BMPs (bone morphogenetic proteins) upon injury to this process. HO was once thought to be a rare occurrence, but recent statistics from the military suggest that as many as 60% of traumatic injuries, resulting from bomb blasts, have associated HO. In this study, we attempt to define the role of peripheral nerves in this process. Since BMP2 has been shown previously to induce release of the neuroinflammatory molecules, substance P (SP) and calcitonin gene related peptide (CGRP), from peripheral, sensory neurons, we examined this process in vivo. SP and CGRP are rapidly expressed upon delivery of BMP2 and remain elevated throughout bone formation. In animals lacking functional sensory neurons (TRPV1(-/-) ), BMP2-mediated increases in SP and CGRP were suppressed as compared to the normal animals, and HO was dramatically inhibited in these deficient mice, suggesting that neuroinflammation plays a functional role. Mast cells, known to be recruited by SP and CGRP, were elevated after BMP2 induction. These mast cells were localized to the nerve structures and underwent degranulation. When degranulation was inhibited using cromolyn, HO was again reduced significantly. Immunohistochemical analysis revealed nerves expressing the stem cell markers nanog and Klf4, as well as the osteoblast marker osterix, after BMP2 induction, in mice treated with cromolyn. The data collectively suggest that BMP2 can act directly on sensory neurons to induce neurogenic inflammation, resulting in nerve remodeling and the migration/release of osteogenic and other stem cells from the nerve. Further, blocking this process significantly reduces HO, suggesting that the stem cell population contributes to bone formation.
Collapse
Affiliation(s)
- Elizabeth Salisbury
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
El-Hoss J, Sullivan K, Cheng T, Yu NYC, Bobyn JD, Peacock L, Mikulec K, Baldock P, Alexander IE, Schindeler A, Little DG. A murine model of neurofibromatosis type 1 tibial pseudarthrosis featuring proliferative fibrous tissue and osteoclast-like cells. J Bone Miner Res 2012; 27:68-78. [PMID: 21956219 DOI: 10.1002/jbmr.528] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 09/14/2011] [Accepted: 09/22/2011] [Indexed: 11/07/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common genetic condition caused by mutations in the NF1 gene. Patients often suffer from tissue-specific lesions associated with local double-inactivation of NF1. In this study, we generated a novel fracture model to investigate the mechanism underlying congenital pseudarthrosis of the tibia (CPT) associated with NF1. We used a Cre-expressing adenovirus (AdCre) to inactivate Nf1 in vitro in cultured osteoprogenitors and osteoblasts, and in vivo in the fracture callus of Nf1(flox/flox) and Nf1(flox/-) mice. The effects of the presence of Nf1(null) cells were extensively examined. Cultured Nf1(null)-committed osteoprogenitors from neonatal calvaria failed to differentiate and express mature osteoblastic markers, even with recombinant bone morphogenetic protein-2 (rhBMP-2) treatment. Similarly, Nf1(null)-inducible osteoprogenitors obtained from Nf1 MyoDnull mouse muscle were also unresponsive to rhBMP-2. In both closed and open fracture models in Nf1(flox/flox) and Nf1(flox/-) mice, local AdCre injection significantly impaired bone healing, with fracture union being <50% that of wild type controls. No significant difference was seen between Nf1(flox/flox) and Nf1(flox/-) mice. Histological analyses showed invasion of the Nf1(null) fractures by fibrous and highly proliferative tissue. Mean amounts of fibrous tissue were increased upward of 10-fold in Nf1(null) fractures and bromodeoxyuridine (BrdU) staining in closed fractures showed increased numbers of proliferating cells. In Nf1(null) fractures, tartrate-resistant acid phosphatase-positive (TRAP+) cells were frequently observed within the fibrous tissue, not lining a bone surface. In summary, we report that local Nf1 deletion in a fracture callus is sufficient to impair bony union and recapitulate histological features of clinical CPT. Cell culture findings support the concept that Nf1 double inactivation impairs early osteoblastic differentiation. This model provides valuable insight into the pathobiology of the disease, and will be helpful for trialing therapeutic compounds.
Collapse
Affiliation(s)
- Jad El-Hoss
- Kids Research Institute, Orthopaedic Research and Biotechnology, Westmead, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Cell-Based Therapies for Spinal Fusion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 760:148-73. [DOI: 10.1007/978-1-4614-4090-1_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
17
|
Sandbo N, Lau A, Kach J, Ngam C, Yau D, Dulin NO. Delayed stress fiber formation mediates pulmonary myofibroblast differentiation in response to TGF-β. Am J Physiol Lung Cell Mol Physiol 2011; 301:L656-66. [PMID: 21856814 DOI: 10.1152/ajplung.00166.2011] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Myofibroblast differentiation induced by transforming growth factor-β (TGF-β) and characterized by de novo expression of smooth muscle (SM)-specific proteins is a key process in wound healing and in the pathogenesis of fibrosis. We have previously shown that TGF-β-induced expression and activation of serum response factor (SRF) is required for this process. In this study, we examined the signaling mechanism for SRF activation by TGF-β as it relates to pulmonary myofibroblast differentiation. TGF-β stimulated a profound, but delayed (18-24 h), activation of Rho kinase and formation of actin stress fibers, which paralleled SM α-actin expression. The translational inhibitor cycloheximide blocked these processes without affecting Smad-dependent gene transcription. Inhibition of Rho kinase by Y-27632 or depolymerization of actin by latrunculin B resulted in inhibition TGF-β-induced SRF activation and SM α-actin expression, having no effect on Smad signaling. Conversely, stabilization of actin stress fibers by jasplakinolide was sufficient to drive these processes in the absence of TGF-β. TGF-β promoted a delayed nuclear accumulation of the SRF coactivator megakaryoblastic leukemia-1 (MKL1)/myocardin-related transcription factor-A, which was inhibited by latrunculin B. Furthermore, TGF-β also induced MKL1 expression, which was inhibited by latrunculin B, by SRF inhibitor CCG-1423, or by SRF knockdown. Together, these data suggest a triphasic model for myofibroblast differentiation in response to TGF-β that involves 1) initial Smad-dependent expression of intermediate signaling molecules driving Rho activation and stress fiber formation, 2) nuclear accumulation of MKL1 and activation of SRF as a result of actin polymerization, and 3) SRF-dependent expression of MKL1, driving further myofibroblast differentiation.
Collapse
Affiliation(s)
- Nathan Sandbo
- Univ. of Wisconsin School of Medicine and Public Health, 5229 MFCB 1685 Highland Ave, Madison, WI 53705, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Olabisi RM, Lazard Z, Heggeness MH, Moran KM, Hipp JA, Dewan AK, Davis AR, West JL, Olmsted-Davis EA. An injectable method for noninvasive spine fusion. Spine J 2011; 11:545-56. [PMID: 21292563 PMCID: PMC3327508 DOI: 10.1016/j.spinee.2010.12.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 12/01/2010] [Accepted: 12/17/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Bone morphogenetic proteins (BMPs) induce bone formation but are difficult to localize, and subsequent diffusion from the site of interest and short half-life reduce the efficacy of the protein. Currently, spine fusion requires stripping, decortications of the transverse processes, and an autograft harvest procedure. Even in combination with BMPs, clinical spinal fusion has a high failure rate, presumably because of difficulties in localizing sufficient levels of BMP. PURPOSE The goal was to achieve reliable spine fusion through a single injection of a cell-based gene therapy system without the need for any surgical intervention. STUDY DESIGN Eighty-seven immunodeficient (n=44) and immune-competent (n=43) mice were injected along the paraspinous musculature to achieve rapid induction of heterotopic ossification (HO) and ultimately spinal arthrodesis. METHODS Immunodeficient and immune-competent mice were injected with fibroblasts, transduced with an adenoviral vector to express BMP2, along the paraspinous musculature. Bone formation was evaluated via radiographs, microcomputed tomography, and biomechanical analysis. RESULTS ew bridging bone between the vertebrae and the fusion to adjacent skeletal bone was obtained as early as 2 weeks. Reduction in spine flexion-extension also occurred as early as 2 weeks after injection of the gene therapy system, with greater than 90% fusion by 4 weeks in all animals regardless of their genetic background. CONCLUSIONS Injection of our cell-based system into the paraspinous musculature induces spinal fusion that is dependent neither on the cell type nor on the immune status. These studies are the first to harness HO in an immune-competent model as a noninvasive injectable system for clinically relevant spinal fusion and may one day impact human spinal arthrodesis.
Collapse
Affiliation(s)
- Ronke M. Olabisi
- Department of Bioengineering, Rice University, MS 142, 6100 Main St, Houston, TX 77005, USA
| | - ZaWaunyka Lazard
- Center for Cell and Gene Therapy, Baylor College of Medicine, Alkek Graduate School BCMN-N1010, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael H. Heggeness
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - Kevin M. Moran
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - John A. Hipp
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - Ashvin K. Dewan
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - Alan R. Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Alkek Graduate School BCMN-N1010, One Baylor Plaza, Houston, TX 77030, USA,Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer L. West
- Department of Bioengineering, Rice University, MS 142, 6100 Main St, Houston, TX 77005, USA
| | - Elizabeth A. Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Alkek Graduate School BCMN-N1010, One Baylor Plaza, Houston, TX 77030, USA,Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Corresponding author. Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, MS BCM505, Houston, TX 77030, USA. Tel.: (713) 798-1253; fax: (713) 798-1230. (E.A. Olmsted-Davis)
| |
Collapse
|
19
|
Li J, Huang X, Xu X, Mayo J, Bringas P, Jiang R, Wang S, Chai Y. SMAD4-mediated WNT signaling controls the fate of cranial neural crest cells during tooth morphogenesis. Development 2011; 138:1977-89. [PMID: 21490069 DOI: 10.1242/dev.061341] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
TGFβ/BMP signaling regulates the fate of multipotential cranial neural crest (CNC) cells during tooth and jawbone formation as these cells differentiate into odontoblasts and osteoblasts, respectively. The functional significance of SMAD4, the common mediator of TGFβ/BMP signaling, in regulating the fate of CNC cells remains unclear. In this study, we investigated the mechanism of SMAD4 in regulating the fate of CNC-derived dental mesenchymal cells through tissue-specific inactivation of Smad4. Ablation of Smad4 results in defects in odontoblast differentiation and dentin formation. Moreover, ectopic bone-like structures replaced normal dentin in the teeth of Osr2-IresCre;Smad4(fl/fl) mice. Despite the lack of dentin, enamel formation appeared unaffected in Osr2-IresCre;Smad4(fl/fl) mice, challenging the paradigm that the initiation of enamel development depends on normal dentin formation. At the molecular level, loss of Smad4 results in downregulation of the WNT pathway inhibitors Dkk1 and Sfrp1 and in the upregulation of canonical WNT signaling, including increased β-catenin activity. More importantly, inhibition of the upregulated canonical WNT pathway in Osr2-IresCre;Smad4(fl/fl) dental mesenchyme in vitro partially rescued the CNC cell fate change. Taken together, our study demonstrates that SMAD4 plays a crucial role in regulating the interplay between TGFβ/BMP and WNT signaling to ensure the proper CNC cell fate decision during organogenesis.
Collapse
Affiliation(s)
- Jingyuan Li
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Olabisi RM, Lazard ZW, Franco CL, Hall MA, Kwon SK, Sevick-Muraca EM, Hipp JA, Davis AR, Olmsted-Davis EA, West JL. Hydrogel microsphere encapsulation of a cell-based gene therapy system increases cell survival of injected cells, transgene expression, and bone volume in a model of heterotopic ossification. Tissue Eng Part A 2010; 16:3727-36. [PMID: 20673027 DOI: 10.1089/ten.tea.2010.0234] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are well known for their osteoinductive activity, yet harnessing this capacity remains a high-priority research focus. We present a novel technology that delivers high BMP-2 levels at targeted locations for rapid endochondral bone formation, enhancing our preexisting cell-based gene therapy system by microencapsulating adenovirus-transduced cells in nondegradable poly(ethylene glycol) diacrylate (PEGDA) hydrogels before intramuscular delivery. This study evaluates the in vitro and in vivo viability, gene expression, and bone formation from transgenic fibroblasts encapsulated in PEGDA microspheres. Fluorescent viability and cytotoxicity assays demonstrated >95% viability in microencapsulated cells. ELISA and alkaline phosphatase assays established that BMP-2 secretion and specific activity from microencapsulated AdBMP2-transduced fibroblasts were not statistically different from monolayer. Longitudinal transgene expression studies of AdDsRed-transduced fibroblasts, followed through live animal optical fluorescent imaging, showed that microencapsulated cells expressed longer than unencapsulated cells. When comparable numbers of microencapsulated AdBMP2-transduced cells were intramuscularly injected into mice, microcomputed tomography evaluation demonstrated that the resultant heterotopic bone formation was approximately twice the volume of unencapsulated cells. The data suggest that microencapsulation protects cells and prolongs and spatially distributes transgene expression. Thus, incorporation of PEGDA hydrogels significantly advances current gene therapy bone repair approaches.
Collapse
Affiliation(s)
- Ronke M Olabisi
- Department of Bioengineering, Rice University, Houston, Texas 77005, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wang W, Nyman JS, Moss HE, Gutierrez G, Mundy GR, Yang X, Elefteriou F. Local low-dose lovastatin delivery improves the bone-healing defect caused by Nf1 loss of function in osteoblasts. J Bone Miner Res 2010; 25:1658-67. [PMID: 20200958 PMCID: PMC3154004 DOI: 10.1002/jbmr.42] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Postfracture tibial nonunion (pseudoarthrosis) leads to lifelong disability in patients with neurofibromatosis type I (NF1), a disorder caused by mutations in the NF1 gene. To determine the contribution of NF1 in bone healing, we assessed bone healing in the Nf1(ob) (-/-) conditional mouse model lacking Nf1 specifically in osteoblasts. A closed distal tibia fracture protocol and a longitudinal study design were used. During the 21- to 28-day postfracture period, callus volume, as expected, decreased in wild-type but not in Nf1(ob) (-/-) mice, suggesting delayed healing. At these two time points, bone volume (BV/TV) and volumetric bone mineral density (vBMD) measured by 3D micro-computed tomography were decreased in Nf1(ob) (-/-) callus-bridging cortices and trabecular compartments compared with wild-type controls. Histomorphometric analyses revealed the presence of cartilaginous remnants, a high amount of osteoid, and increased osteoclast surfaces in Nf1(ob) (-/-) calluses 21 days after fracture, which was accompanied by increased expression of osteopontin, Rankl, and Tgfbeta. Callus strength measured by three-point bending 28 days after fracture was reduced in Nf1(ob) (-/-) versus wild-type calluses. Importantly, from a clinical point of view, this defect of callus maturation and strength could be ameliorated by local delivery of low-dose lovastatin microparticles, which successfully decreased osteoid volume and cartilaginous remnant number and increased callus BV/TV and strength in mutant mice. These results thus indicate that the dysfunctions caused by loss of Nf1 in osteoblasts impair callus maturation and weaken callus mechanical properties and suggest that local delivery of low-dose lovastatin may improve bone healing in NF1 patients.
Collapse
Affiliation(s)
- Weixi Wang
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN 37232-0575, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Heterotopic ossification (HO), or endochondral bone formation at nonskeletal sites, often results from traumatic injury and can lead to devastating consequences. Alternatively, the ability to harness this phenomenon would greatly enhance current orthopedic tools for treating segmental bone defects. Thus, understanding the earliest events in this process potentially would allow us to design more targeted therapies to either block or enhance this process. Using a murine model of HO induced by delivery of adenovirus-transduced cells expressing bone morphogenetic protein 2 (BMP-2), we show here that one of the earliest stages in this process is the establishment of new vessels prior to the appearance of cartilage. As early as 48 hours after induction of HO, we observed the appearance of brown adipocytes expressing vascular endothelial growth factors (VEGFs) simultaneous with endothelial progenitor replication. This was determined by using a murine model that possesses the VEGF receptor 2 (Flk1) promoter containing an endothelial cell enhancer driving the expression of nuclear-localized yellow fluorescent protein (YFP). Expression of this marker has been shown previously to correlate with the establishment of new vasculature, and the nuclear localization of YFP expression allowed us to quantify changes in endothelial cell numbers. We found a significant increase in Flk1-H2B::YFP cells in BMP-2-treated animals compared with controls. The increase in endothelial progenitors occurred 3 days prior to the appearance of early cartilage. The data collectively suggest that vascular remodeling and growth may be essential to modify the microenvironment and enable engraftment of the necessary progenitors to form endochondral bone.
Collapse
|
23
|
Nakashima H, Kaur B, Chiocca EA. Directing systemic oncolytic viral delivery to tumors via carrier cells. Cytokine Growth Factor Rev 2010; 21:119-26. [PMID: 20226717 DOI: 10.1016/j.cytogfr.2010.02.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The systemic administration of oncolytic virus (OV) is often inefficient due to clearance of the virus by host defense mechanism and spurious targeting of non-cancer tissues through the bloodstream. Cell mediated OV delivery could hide the virus from host defenses and direct them toward tumors: Mesenchymal and neural stem cells have been described to possess tumor-homing ability as well as the capacity to deliver OVs. In this review, we will focus on approaches where OV and carrier cells are utilized for cancer therapy. Effective cellular internalization and replication of OVs need to occur both in cancer and carrier cells. We thus will discuss the current challenges faced by the use of OV delivery via carrier cells.
Collapse
Affiliation(s)
- Hiroshi Nakashima
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center, Columbus, OH 43210, United States
| | | | | |
Collapse
|
24
|
Fernandez-Fuente M, Ames EG, Wagner ML, Zhou H, Strom M, Zammit PS, Mickelson JR, Muntoni F, Brown SC, Piercy RJ. Assessment of the transformation of equine skin-derived fibroblasts to multinucleated skeletal myotubes following lentiviral-induced expression of equine myogenic differentiation 1. Am J Vet Res 2009; 69:1637-45. [PMID: 19046012 DOI: 10.2460/ajvr.69.12.1637] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To develop a reliable method for converting cultured equine skin-derived fibroblasts into muscle cells. SAMPLE POPULATION Equine skin-derived fibroblasts. PROCEDURES The equine myogenic differentiation 1 (eqMyoD) genomic sequence was obtained by use of equine bacterial artificial chromosome screening and PCR sequencing. Total mRNA was extracted from foal skeletal muscle, and eqMyoD cDNA was cloned into a plasmid vector with an internal ribosomal entry site to express bicistronic eqMyoD or enhanced green fluorescent protein (EGFP). Transient expression was confirmed by immunocytochemical analysis and western immunoblots in equine fibroblasts and fibroblasts from National Institutes of Health Swiss mouse embryos, prior to generation of a lentiviral vector containing the same coding sequences. Transformation of equine skin-derived cells into skeletal myotubes was examined by use of immunohistochemical analysis, western immunoblotting, and periodic acid-Schiff staining. RESULTS eqMyoD mRNA consists of 960 bp and shares high homology with myogenic differentiation 1 from other mammals. Transfection confirmed the expression of a 53-kd protein with mainly nuclear localization. Lentiviral transduction was efficient, with approximately 80% of EGFP-positive cells transformed into multinucleated myotubes during 15 days, as determined by expression of the muscle-specific proteins desmin, troponin-T, and sarcomeric myosin and by cytoplasmic storage of glycogen. CONCLUSIONS AND CLINICAL RELEVANCE Equine primary fibroblasts were transformed by lentiviral transduction of eqMyoD into fusion-competent myoblasts. This may offer a preferable alternative to primary myoblast cultures for the investigation of cellular defects associated with muscle diseases of horses, such as recurrent exertional rhabdomyolysis and polysaccharide storage myopathy.
Collapse
Affiliation(s)
- Marta Fernandez-Fuente
- Comparative Neuromuscular Diseases Laboratory, Department of Veterinary Clinical Sciences, Royal Veterinary College, Hawkshead Ln, Hertfordshire AL9 7TA, England
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Physicochemical properties of gene transfer vectors play an important role in both transduction efficiency and biodistribution following airway delivery. Adeno-associated virus (AAV) vectors are currently used in many gene transfer applications; however, the respiratory epithelium remains a challenging target. We synthesized two cationic sterol-based lipids, dexamethasone-spermine (DS) and disubstituted spermine (D(2)S) for pulmonary gene targeting. Scanning and transmission electron micrographs (TEM) confirmed that AAV/lipid formulations produced submicron-sized clusters. When AAV2/9 or AAV2/6.2 were formulated with these cationic lipids, the complexes had positive zeta potential (zeta) and the transduction efficiency in cultured A549 cells increased by sevenfold and sixfold, respectively. Transduction of cultured human airway epithelium with AAV2/6.2-lipid formulations also showed approximately twofold increase in green fluorescence protein (GFP) positive cells as quantified by flow cytometry. Intranasal administration of 10(11) genome copies (GC) of AAV2/9 and AAV2/6.2 coformulated with lipid formulations resulted in an average fourfold increase in transgene expression for both vectors. Formulation of AAV2/9 with DS changed the tropism of this vector for the alveolar epithelium, resulting in successful transduction of conducting airway epithelium. Our results suggest that formulating AAV2/9 and AAV2/6.2 with DS and D(2)S can lead to improved physicochemical characteristics for in vivo gene delivery to lung.
Collapse
|
26
|
Evans CH, Ghivizzani SC, Robbins PD. Orthopedic gene therapy in 2008. Mol Ther 2009; 17:231-44. [PMID: 19066598 PMCID: PMC2835052 DOI: 10.1038/mt.2008.265] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 10/26/2008] [Indexed: 02/07/2023] Open
Abstract
Orthopedic disorders, although rarely fatal, are the leading cause of morbidity and impose a huge socioeconomic burden. Their prevalence will increase dramatically as populations age and gain weight. Many orthopedic conditions are difficult to treat by conventional means; however, they are good candidates for gene therapy. Clinical trials have already been initiated for arthritis and the aseptic loosening of prosthetic joints, and the development of bone-healing applications is at an advanced, preclinical stage. Other potential uses include the treatment of Mendelian diseases and orthopedic tumors, as well as the repair and regeneration of cartilage, ligaments, and tendons. Many of these goals should be achievable with existing technologies. The main barriers to clinical application are funding and regulatory issues, which in turn reflect major safety concerns and the opinion, in some quarters, that gene therapy should not be applied to nonlethal, nongenetic diseases. For some indications, advances in nongenetic treatments have also diminished enthusiasm. Nevertheless, the preclinical and early clinical data are impressive and provide considerable optimism that gene therapy will provide straightforward, effective solutions to the clinical management of several common debilitating disorders that are otherwise difficult and expensive to treat.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopaedics, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
27
|
Abstract
Over the past three decades many techniques for expressing exogenous genes in a variety of cells and cell lines have been developed. Exogenous gene expression in macrophages has lagged behind that of other nonhematopioetic cells. There are many reasons for this, but most are due to technical difficulties associated with transfecting macrophages. As professional phagocytes, macrophages are endowed with many potent degradative enzymes that can disrupt nucleic acid integrity and make gene transfer into these cells an inefficient process. This is especially true of activated macrophages which undergo a dramatic change in their physiology following exposure to immune or inflammatory stimuli. Viral transduction of these cells has been hampered because macrophages are end-stage cells that generally do not divide; therefore, some of the vectors that depend on integration into a replicative genome have met with limited success. Furthermore, macrophages are quite responsive to "danger signals," and therefore several of the original viral vectors that were used for gene transfer induced potent anti-viral responses in these cells making these vectors inappropriate for gene delivery. Many of these difficulties have been largely overcome, and relatively high efficiency gene expression in primary human or murine macrophages is becoming more routine. In the present chapter we discuss some of the gene expression techniques that have met with success and review the advantages and disadvantages of each.
Collapse
|
28
|
A simple, lanthanide-based method to enhance the transduction efficiency of adenovirus vectors. Gene Ther 2008; 15:357-63. [PMID: 18283289 DOI: 10.1038/sj.gt.3303092] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Based upon the powerful bridging and charge-masking properties of lanthanide cations (Ln3+), we have investigated their use to improve the transduction efficiency of adenovirus vectors. Using a luciferase marker gene, it was possible to increase transgene expression by the murine mesenchymal stem cell line C3H10T(1/2) by up to four log orders when using very low multiplicities of infection in conjunction with Ln3+; La3+ was superior to Gd3+, Y3+ and Lu3+ in this regard. All Ln3+ were more effective than Ca2+. Flow cytometry, using a green fluorescent protein marker gene, confirmed that La3+ increased both the percentage of transduced cells and the level of transgene expression per cell. Transduction of primary cultures of a variety of different mesenchymal cells from human, rabbit, bovine and rat sources, as well as gene transfer to synovium and muscle in vivo, was also greatly enhanced. Our findings suggest that this lanthanide-based method holds much promise for expediting both experimental and clinical applications of gene transfer with adenoviral vectors.
Collapse
|
29
|
Fouletier-Dilling CM, Gannon FH, Olmsted-Davis EA, Lazard Z, Heggeness MH, Shafer JA, Hipp JA, Davis AR. Efficient and rapid osteoinduction in an immune-competent host. Hum Gene Ther 2007; 18:733-45. [PMID: 17691858 DOI: 10.1089/hum.2006.190] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Osteoinductive systems to induce targeted rapid bone formation hold clinical promise, but development of technologies for clinical use that must be tested in animal models is often a difficult challenge. We previously demonstrated that implantation of human cells transduced with Ad5F35BMP2 to express high levels of bone morphogenetic protein-2 (BMP2) resulted in rapid bone formation at targeted sites. Inclusion of human cells in this model precluded us from testing this system in an immune-competent animal model, thus limiting information about the efficacy of this approach. Here, for the first time we demonstrate the similarity between BMP2-induced endochondral bone formation in a system using human cells in an immune-incompetent mouse and a murine cell-based BMP2 gene therapy system in immune-competent animals. In both cases the delivery cells are rapidly cleared, within 5 days, and in neither case do they appear to contribute to any of the structures forming in the tissues. Endochondral bone formation progressed through a highly ordered series of stages that were both morphologically and temporally indistinguishable between the two models. Even longterm analysis of the heterotopic bone demonstrated similar bone volumes and the eventual remodeling to form similar structures. The results suggest that the ability of BMP2 to rapidly induce bone formation overrides contributions from either immune status or the nature of delivery cells.
Collapse
Affiliation(s)
- Christine M Fouletier-Dilling
- Center for Cell and Gene Therapy, Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Shafer J, Davis AR, Gannon FH, Fouletier-Dilling CM, Lazard Z, Moran K, Gugala Z, Ozen M, Ittmann M, Heggeness MH, Olmsted-Davis E. Oxygen Tension Directs Chondrogenic Differentiation of Myelo-Monocytic Progenitors During Endochondral Bone Formation. ACTA ACUST UNITED AC 2007; 13:2011-9. [PMID: 17518751 DOI: 10.1089/ten.2006.0063] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Synthesis of bone requires both essential progenitors to form the various structures and the correct microenvironment for their differentiation. To identify these factors, we have used a system that exploits bone morphogenetic protein's ability to induce endochondral bone formation rapidly. One of the earliest events observed was the influx and proliferation of fibroblastic cells that express both vascular smooth muscle cell markers, alpha smooth muscle actin (alpha SMA), smooth muscle myosin heavy chain, and the monocytic marker CD68. The expression of these factors was lost by days 4 to 5, coincident with the up-regulation of Sox9 and the appearance of chondrocytes. Studies with a cyclization recombination (Cre)/lox system, in which a myeloid-specific promoter driving Cre recombinase can irreversibly unblock expression of beta-galactosidase only in cells of myeloid origin, showed specific activity in the newly formed chondrocytes. These results suggest that early chondrocyte progenitors are of myeloid origin. Simultaneous with this recruitment, we determined that a numbers of these cells were in a hypoxic state, indicative of a low-oxygen environment. The cells in the hypoxic regions were undergoing chondrogenesis, whereas cells in adjacent normoxic regions appeared to be assembling into new vessels, suggesting that the oxygen microenvironment is critical for establishment of the cartilage.
Collapse
Affiliation(s)
- Jessica Shafer
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Olmsted-Davis E, Gannon FH, Ozen M, Ittmann MM, Gugala Z, Hipp JA, Moran KM, Fouletier-Dilling CM, Schumara-Martin S, Lindsey RW, Heggeness MH, Brenner MK, Davis AR. Hypoxic adipocytes pattern early heterotopic bone formation. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:620-32. [PMID: 17255330 PMCID: PMC1851874 DOI: 10.2353/ajpath.2007.060692] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The factors contributing to heterotopic ossification, the formation of bone in abnormal soft-tissue locations, are beginning to emerge, but little is known about microenvironmental conditions promoting this often devastating disease. Using a murine model in which endochondral bone formation is triggered in muscle by bone morphogenetic protein 2 (BMP2), we studied changes near the site of injection of BMP2-expressing cells. As early as 24 hours later, brown adipocytes began accumulating in the lesional area. These cells stained positively for pimonidazole and therefore generated hypoxic stress within the target tissue, a prerequisite for the differentiation of stem cells to chondrocytes and subsequent heterotopic bone formation. We propose that aberrant expression of BMPs in soft tissue stimulates production of brown adipocytes, which drive the early steps of heterotopic endochondral ossification by lowering oxygen tension in adjacent tissue, creating the correct environment for chondrogenesis. Results in misty gray lean mutant mice not producing brown fat suggest that white adipocytes convert into fat-oxidizing cells when brown adipocytes are unavailable, providing a compensatory mechanism for generation of a hypoxic microenvironment. Manipulation of the transcriptional control of adipocyte fate in local soft-tissue environments may offer a means to prevent or treat development of bone in extraskeletal sites.
Collapse
MESH Headings
- Adipocytes, Brown/metabolism
- Adipocytes, Brown/pathology
- Adipocytes, Brown/transplantation
- Animals
- Bone Morphogenetic Protein 2
- Bone Morphogenetic Proteins/biosynthesis
- Cell Differentiation
- Cell Hypoxia/genetics
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Chondrogenesis
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Ossification, Heterotopic/genetics
- Ossification, Heterotopic/metabolism
- Ossification, Heterotopic/pathology
- Ossification, Heterotopic/therapy
- Stem Cells/metabolism
- Stem Cells/pathology
- Transforming Growth Factor beta/biosynthesis
Collapse
Affiliation(s)
- Elizabeth Olmsted-Davis
- Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Intact or genetically manipulated mesnechymal stem cells (MSCs) are being considered an important cell source for developing human cell-based therapeutic approaches. For applications in which transient, high-level expression of the transgene is necessary, adenovirus vectors have become increasingly popular gene-transfer vehicles. However, host range and cell-type tropism restrict the use of specific adenovectors, sometimes necessitating the lengthy development of vectors with appropriate cell specificity. Here, we present a versatile and inexpensive porcine MSC transduction procedure that can also be used on other cell types from various species, including human that are otherwise refractory to adenovirus infection.
Collapse
Affiliation(s)
- Pablo Bosch
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | | |
Collapse
|
33
|
Bosch P, Fouletier-Dilling C, Olmsted-Davis EA, Davis AR, Stice SL. Efficient adenoviral-mediated gene delivery into porcine mesenchymal stem cells. Mol Reprod Dev 2006; 73:1393-403. [PMID: 16897738 DOI: 10.1002/mrd.20593] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Mesenchymal stem cell (MSC) mediated gene therapy research has been conducted predominantly on rodents. Appropriate large animal models may provide additional safety and efficacy information prior to human clinical trials. The objectives of this study were: (a) to optimize adenoviral transduction efficiency of porcine bone marrow MSCs using a commercial polyamine-based transfection reagent (GeneJammer, Stratagene, La Jolla, CA), and (b) to determine whether transduced MSCs retain the ability to differentiate into mesodermal lineages. Porcine MSCs (pMSCs) were infected under varying conditions, with replication-defective adenoviral vectors carrying the GFP gene and GFP expression analyzed. Transduced cells were induced to differentiate in vitro into adipogenic, chondrogenic, and osteogenic lineages. We observed a 5.5-fold increase in the percentage of GFP-expressing pMSCs when adenovirus type 5 carrying the adenovirus type 35 fiber (Ad5F35eGFP) was used in conjunction with GeneJammer. Transduction of pMSCs at 10.3-13.8 MOI (1,500-2,000 vp/cell) in the presence of Gene Jammer yielded the highest percentage of GFP-expressing cells ( approximately 90%) without affecting cell viability. A similar positive effect was detected when pMSCs were infected with an Ad5eGFP vector. Presence of fetal bovine serum (FBS) during adenoviral transduction enhanced vector-encoded transgene expression in both GeneJammer-treated and control groups. pMSCs transduced with adenovirus vector in the presence of GeneJammer underwent lipogenic, chondrogenic, and osteogenic differentiation. Addition of GeneJammer during adenoviral infection of pMSCs can revert the poor transduction efficiency of pMSCs while retaining their pluripotent differentiation capacity. GeneJammer-enhanced transduction will facilitate the use of adenoviral vectors in MSC-mediated gene therapy models and therapies.
Collapse
Affiliation(s)
- P Bosch
- Department of Animal and Dairy Science, The University of Georgia, Athens, Georgia 30602, USA
| | | | | | | | | |
Collapse
|