Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 16225, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29176755

ABSTRACT

Osteoporotic patients often suffer from bone fracture but its healing is compromised due to impaired osteogenesis potential of bone marrow-derived mesenchymal stem cells (BMSCs). Here we aimed to exploit adipose-derived stem cells from ovariectomized rats (OVX-ASCs) for bone healing. We unraveled that OVX-ASCs highly expressed miR-214 and identified 2 miR-214 targets: CTNNB1 (ß-catenin) and TAB2. We demonstrated that miR-214 targeting of these two genes blocked the Wnt pathway, led to preferable adipogenesis and hindered osteogenesis. As a result, OVX-ASCs implantation into OVX rats failed to heal critical-size metaphyseal bone defects. We further engineered the OVX-ASCs with a novel Cre/loxP-based hybrid baculovirus vector that conferred prolonged expression of miR-214 sponge. Gene delivery for miR-214 sponge expression successfully downregulated miR-214 levels, activated the Wnt pathway, upregulated osteogenic factors ß-catenin/Runx2, downregulated adipogenic factors PPAR-γ and C/EBP-α, shifted the differentiation propensity towards osteogenic lineage, enhanced the osteogenesis of co-cultured OVX-BMSCs, elevated BMP7/osteoprotegerin secretion and hindered exosomal miR-214/osteopontin release. Consequently, implanting the miR-214 sponge-expressing OVX-ASCs tremendously improved bone healing in OVX rats. Co-expression of miR-214 sponge and BMP2 further synergized the OVX-ASCs-mediated bone regeneration in OVX rats. This study implicates the potential of suppressing miR-214 by baculovirus-mediated gene delivery in osteoporotic ASCs for regenerative medicine.


Subject(s)
Bone Regeneration , Cell Differentiation , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Osteoporosis, Postmenopausal/metabolism , Adipose Tissue/cytology , Animals , Baculoviridae/genetics , Bone Morphogenetic Protein 7/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cells, Cultured , Female , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Osteoporosis, Postmenopausal/therapy , PPAR gamma/metabolism , RNAi Therapeutics/methods , Rats , Rats, Sprague-Dawley , Sf9 Cells , Spodoptera
2.
Biomaterials ; 140: 189-200, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28658635

ABSTRACT

Peripheral nerve regeneration requires coordinated functions of supporting cells (e.g. Schwann cells) and neurotrophic factors such as glial cell line-derived neurotrophic factor (GDNF), but nerve regeneration is usually far from complete. Here we constructed a Cre/loxP-based hybrid baculovirus (BV) vector which enabled intracellular formation of episomal DNA minicircle for effective transduction of rat adipose-derived stem cells (ASCs) and prolonged expression of functional GDNF capable of recruiting Schwann cells. The GDNF expression persisted for >20 days with the peak level (≈128 ng/ml) tremendously exceeding the picogram levels of GDNF secreted by neuroprogenitor cells. We further developed a facile method to fabricate and transduce cell sheets composed of undifferentiated ASCs in 2 days, without the need of thermo-responsive polymer commonly used for cell sheet fabrication. Implantation of the hybrid BV-engineered, GDNF-expressing ASCs sheets into sciatic nerve transection site in rats significantly improved the nerve repair, as judged from the enhanced functional recovery, nerve reinnervation, electrophysiological functionality, Schwann cells proliferation/infiltration, axon regeneration, myelination and angiogenesis. The hybrid BV is able to functionalize ASCs sheets by intracellular episomal DNA minicircle formation that circumvents undesired gene integration, and the ASCs sheets fabrication is rapid and simple. These data and features implicate the potentials of ASCs sheets functionalized by the hybrid BV for peripheral nerve regeneration.


Subject(s)
Adipose Tissue/cytology , Baculoviridae/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Nerve Regeneration , Sciatic Nerve/physiology , Sciatic Neuropathy/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Cells, Cultured , Female , Humans , Male , Rats, Sprague-Dawley , Recovery of Function , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology , Stem Cells/metabolism , Transduction, Genetic
3.
Biomaterials ; 124: 1-11, 2017 04.
Article in English | MEDLINE | ID: mdl-28182872

ABSTRACT

Calvarial bone repair remains challenging for adults. Although adipose-derived stem cells (ASCs) hold promise to heal bone defects, use of ASCs for critical-size calvarial bone repair is ineffective. Stromal cell-derived factor 1 (SDF-1) is a chemokine capable of triggering stem cell migration. Although recombinant SDF-1 protein is co-delivered with other molecules including BMP-2 to facilitate calvarial bone repair, these approaches did not yield satisfactory healing. This study aimed to exploit a newly developed Cre/loxP-based hybrid baculovirus for efficient gene delivery and prolonged transgene expression in ASCs. We demonstrated that transduction of rat ASCs with the hybrid Cre/loxP-based baculovirus enabled robust and sustained expression of functional BMP-2 and SDF-1. Expression of BMP-2 or SDF-1 alone failed to effectively induce rat ASCs osteogenesis and healing of critical-size calvarial bone defects. Nonetheless, prolonged BMP-2/SDF-1 co-expression in ASCs synergistically activated both Smad and ERK1/2 pathways and hence potentiated the osteogenesis. Consequently, transplantation of the hybrid baculovirus-engineered, BMP-2/SDF-1-expressing ASCs/scaffold constructs potently healed the critical-size (6 mm) calvarial bone defects (filling ≈70% of defect volume), which considerably outperformed the calvarial bone repair using BMP-2/SDF-1 delivered with biomaterial-based scaffolds. These data implicated the potential of Cre/loxP-based hybrid baculovirus vector for ASCs engineering and calvarial bone healing.


Subject(s)
Adult Stem Cells/physiology , Adult Stem Cells/transplantation , Bone Morphogenetic Protein 2/metabolism , Cell Engineering/methods , Chemokine CXCL12/metabolism , Skull Fractures/therapy , Transduction, Genetic/methods , Adult Stem Cells/virology , Animals , Baculoviridae/genetics , Bone Morphogenetic Protein 2/genetics , Bone Regeneration/physiology , Cells, Cultured , Chemokine CXCL12/genetics , Female , Rats , Rats, Sprague-Dawley , Skull Fractures/pathology , Skull Fractures/physiopathology , Treatment Outcome
4.
J Tissue Eng Regen Med ; 11(11): 3068-3077, 2017 11.
Article in English | MEDLINE | ID: mdl-27687795

ABSTRACT

Repairing large calvarial bone defects remains a challenging task. Previously, it was discovered that that miR-148b, when acting in concert with bone morphogenetic protein 2 (BMP-2), enhanced the osteogenesis of human adipose-derived stem cells (hASCs) and improved calvarial bone healing in nude mice. However, the molecular target of miR-148b remained elusive. Here it is revealed that miR-148b directly targets NOG, whose gene product (noggin) is an antagonist to BMPs and negatively regulates BMP-induced osteogenic differentiation and bone formation. A new Cre/loxP-based baculovirus system was employed to drive prolonged BMP-2 and miR-148b overexpression in hASCs, wherein the BMP-2 overexpression induced noggin expression but the concurrent miR-148b expression downregulated noggin, thus relieving the negative regulatory loop and ameliorating hASC osteogenesis without hindering hASC proliferation or triggering appreciable cytotoxicity. Implantation of the engineered hASCs coexpressing BMP-2 and miR-148b into nude mice enabled substantial repair of critical-size calvarial bone defects (4 mm diameter) at 12 weeks post-transplantation, filling 83% of the defect area, 75% of bone volume and restoring the bone density to 89% of the original bone density. Such superior healing effects indicate the potential of the Cre/loxP-based baculovirus-mediated BMP-2/miR-148b expression for calvarial bone repair. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Adipose Tissue/metabolism , Bone Morphogenetic Protein 2/biosynthesis , Bone Regeneration , Gene Expression , MicroRNAs/biosynthesis , Skull , Stem Cell Transplantation , Stem Cells/metabolism , Adipose Tissue/pathology , Animals , Baculoviridae , Bone Morphogenetic Protein 2/genetics , Female , Heterografts , Humans , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Skull/injuries , Skull/metabolism , Skull/pathology , Stem Cells/pathology , Transduction, Genetic
5.
ACS Appl Mater Interfaces ; 8(19): 12082-90, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27111420

ABSTRACT

Metal nanowires are promising for their applications including electrical connectors, transparent conductive electrodes and conductive additives, but the use of metal nanowires as photothermal agents to convert light to heat has yet to be reported. Here we synthesized dispersible polyethylene glycol-coated (PEGylated) copper nanowires (CuNWs) and showed for the first time that PEGylated CuNWs were able to convert near-infrared (NIR, 808 nm) light into heat at a photothermal efficiency of 12.5%. The PEGylated CuNWs exhibited good reusability and enabled rapid temperature rise to >50 °C in 6 min by NIR irradiation. The PEGylated CuNWs were flexible and intertwined around the cancer cells, which, upon NIR irradiation, allowed for direct heat transmission to cells and effectively triggered cancer cell ablation in vitro. Intratumoral injection of PEGylated CuNWs into colon tumor-bearing mice and ensuing NIR irradiation for 6 min significantly raised the local temperature to >50 °C, induced necrosis, and suppressed tumor growth. Compared with other NIR light absorbing noble metal-based nanomaterials, PEGylated CuNWs are relatively easy to synthesize in both laboratory and large scales using the low cost copper. This study demonstrated the potential of PEGylated CuNWs as a new cost-effective photothermal agent, and paved a new avenue to using CuNWs for cancer therapy.


Subject(s)
Coated Materials, Biocompatible , Colonic Neoplasms/therapy , Copper , Hyperthermia, Induced/methods , Nanowires , Phototherapy/methods , Polyethylene Glycols , Animals , Cell Line, Tumor , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Copper/chemistry , Copper/pharmacology , Female , Mice , Mice, Inbred BALB C , Nanowires/chemistry , Nanowires/therapeutic use , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology
6.
Biomaterials ; 74: 155-66, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26454414

ABSTRACT

Fractures associated with osteoporosis are a worldwide health problem. To augment osteoporotic bone healing, we aimed to develop a cell/gene therapy approach in combination with miRNA manipulation. We unraveled aberrant overexpression of miR-140* and miR-214 in the bone marrow-derived MSCs isolated from ovariectomized (OVX) rats (OVX-BMSCs). To suppress the miRNA levels, we constructed hybrid baculovirus vectors expressing miRNA sponges to antagonize miR-140* or miR-214. Engineering OVX-BMSCs with the hybrid vectors persistently attenuated the cellular miR-140*/miR-214 levels, which promoted the OVX-BMSCs osteogenesis and augmented the ability of OVX-BMSCs to repress osteoclast maturation in vitro. Notably, suppressing miR-214 exerted more potent osteoinductive effects. In the osteoporotic rat models with a critical-size bone defect at the femoral metaphysis, implanting the OVX-BMSCs ectopically expressing BMP2 failed to heal the defect, which underscored the difficulty to heal osteoporotic bone defects. Nonetheless, allotransplantation of the miR-214 sponges-expressing OVX-BMSCs healed the defect and ameliorated the bone quality (density, trabecular number, trabecular thickness and trabecular space) at 4 weeks post-implantation. Co-expressing BMP2 and miR-214 sponges in OVX-BMSCs further synergistically substantiated the healing. The baculovirus-engineered OVX-BMSCs that expressed miR-214 sponge, with or without BMP2 expression, thus paved a new avenue to the treatment of osteoporotic bone defects.


Subject(s)
Baculoviridae/genetics , Bone Marrow Cells/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Osteoporosis/pathology , Animals , Fracture Healing , Ovariectomy , Rats , X-Ray Microtomography
7.
Biomaterials ; 50: 98-106, 2015 May.
Article in English | MEDLINE | ID: mdl-25736500

ABSTRACT

Adipose-derived stem cells (ASCs) hold promise for bone regeneration but possess inferior osteogenesis potential. Allotransplantation of ASCs engineered with the BMP2/VEGF-expressing baculoviruses into rabbits healed critical-size segmental bone defects. To translate the technology to clinical applications, we aimed to demonstrate massive bone healing in minipigs that more closely mimicked the clinical scenarios, using a new hybrid baculovirus system consisting of BacFLPo expressing the codon-optimized FLP recombinase (FLPo) and the substrate baculovirus harboring the transgene flanked by Frt sequences. Co-transduction of minipig ASCs (pASCs) with BacFLPo and the substrate baculovirus enabled transgene cassette excision, recombination and minicircle formation in ≈73.7% of pASCs, which substantially prolonged the transgene (BMP2 and VEGF) expression to 28 days. When encoding BMP2, the FLPo/Frt-based system augmented the pASCs osteogenesis. Allotransplantation of the BMP2/VEGF-expressing pASCs into minipigs healed massive segmental bone defects (30 mm in length) at the mid-diaphysis of femora, as evaluated by computed tomography, positron emission tomography, histology, immunohistochemical staining and biochemical testing. The defect size was ≈15% of femoral length in minipigs and was equivalent to ≈60-70 mm of femoral defect in humans, thus the healing using pASCs engineered with the FLPo/Frt-based baculovirus represented a remarkable advance for the treatment of massive bone defects.


Subject(s)
Baculoviridae/metabolism , DNA Nucleotidyltransferases/metabolism , Femur/pathology , Genetic Vectors/metabolism , Stem Cell Transplantation , Stem Cells/cytology , Wound Healing , Adipose Tissue/cytology , Animals , Base Sequence , Biomechanical Phenomena , Bone Regeneration , Cells, Cultured , Femur/blood supply , Femur/diagnostic imaging , Genetic Engineering , Osteogenesis , Positron-Emission Tomography , Stem Cells/metabolism , Swine , Swine, Miniature , Tomography, X-Ray Computed , Transgenes , Transplantation, Homologous
8.
Adv Healthc Mater ; 4(7): 948-68, 2015 May.
Article in English | MEDLINE | ID: mdl-25656682

ABSTRACT

Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.


Subject(s)
Cartilage, Articular/physiology , Gene Regulatory Networks/genetics , Regeneration/genetics , Tissue Engineering/methods , Animals , Genetic Therapy/methods , Humans
9.
Tissue Eng Part A ; 21(9-10): 1471-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25602313

ABSTRACT

We recently developed hybrid baculovirus (BV) vectors that exploited FLPo/Frt-mediated DNA minicircle formation. Engineering of adipose-derived stem cells (ASCs) with the FLPo/Frt-based BV vectors enabled prolonged transgene expression and, after cell implantation into rabbits, ameliorated cartilage regeneration and bone repair. To translate the hybrid BV one step further toward clinical applications, here we assessed the biosafety profiles of the hybrid BV-engineered human ASCs (hASCs) in vitro and evaluated the immune responses elicited by the engineered porcine ASCs (pASCs) in large animals. We confirmed that the hybrid BV did not compromise the hASCs viability, immunosuppressive capacity, and surface characteristics. Neither did the hybrid BV cause chromosomal abnormality/transgene integration in vitro nor did it induce tumorigenicity in vivo. In the large animal study, pASCs were engineered with the hybrid BV expressing bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factor (VEGF) and implanted into femoral bone defects in mini pigs. The hybrid BV-engineered pASCs enabled prolonged BMP2/VEGF expression and triggered the healing of massive segmental bone defects, while only eliciting transient antibody, cytokine, and local cellular immune responses stemming from the implantation procedure itself. These data altogether demonstrated the safety of the hybrid BV vectors for ASCs engineering and bone healing in large animals, hence implicating the potential in clinical applications.


Subject(s)
Adipose Tissue/cytology , Baculoviridae/metabolism , Cell Engineering/methods , Proteins/metabolism , Stem Cells/cytology , Animals , Antibody Formation , Base Sequence , Carcinogenesis/pathology , Cell Line, Tumor , Chromosomes, Human/metabolism , Codon/genetics , Female , Genetic Vectors , Humans , Immunity, Cellular , Inflammation/immunology , Mice, Nude , Models, Animal , Oncogenes , Rabbits , Sus scrofa , Th1 Cells/immunology , Th2 Cells/immunology , Transduction, Genetic , Transgenes
10.
Biomaterials ; 40: 12-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25498801

ABSTRACT

Graphene oxide (GO) is a nanomaterial that provokes autophagy in CT26 colon cancer cells and confers antitumor effects. Here we demonstrated that both GO and the chemotherapy drug cisplatin (CDDP) induced autophagy but elicited low degrees of CT26 cell death. Strikingly, GO combined with CDDP (GO/CDDP) potentiated the CT26 cell killing via necrosis. GO/CDDP not only elicited autophagy, but induced the nuclear import of CDDP and the autophagy marker LC3. The nuclear LC3 did not co-localize with p62 or Lamp-2, neither did blocking autolysosome formation significantly hinder the nuclear import of LC3/CDDP and necrosis, indicating that autophagosome and autolysosome formation was dispensable. Conversely, suppressing phagophore formation and importin-α/ß significantly alleviated the nuclear import of LC3/CDDP and necrosis. These data suggested that GO/CDDP diverted the LC3 flux in the early phase of autophagy, resulting in LC3 trafficking towards the nucleus in an importin-α/ß-dependent manner, which concurred with the CDDP nuclear import and necrosis. Intratumoral injection of GO/CDDP into mice bearing CT26 colon tumors potentiated immune cell infiltration and promoted cell death, autophagy and HMGB1 release, thereby synergistically augmenting the antitumor effects. Altogether, we unveiled a mechanism concerning how nanomaterials chemosensitize cancer cells and demonstrated the potentials of GO as a chemosensitizer.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy , Colonic Neoplasms/drug therapy , Graphite/therapeutic use , Oxides/therapeutic use , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Colonic Neoplasms/pathology , Graphite/pharmacology , Mice, Inbred BALB C , Microtubule-Associated Proteins/metabolism , Necrosis , Oxides/pharmacology
11.
Mol Ther ; 23(1): 79-88, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25023326

ABSTRACT

MicroRNA 122 (miR-122) is a tumor suppressor for hepatocellular carcinoma (HCC) but is lowly expressed in HCC cells. MiR-151 is aberrantly overexpressed in HCC cells and promotes HCC metastasis yet its roles on HCC tumorigenicity are unknown. To combat HCC tumorigenicity/metastasis, we developed Sleeping Beauty (SB)-based hybrid baculovirus (BV) vectors that expressed (i) miR-122 precursors (pre-miR-122), (ii) miR-151 sponges, or (iii) pre-miR-122 and miR-151 sponges. Transduction of aggressive HCC cells (Mahlavu) with the pre-miR-122-expressing BV tremendously enhanced miR-122 levels for >6 weeks, suppressed the levels of downstream effectors (e.g., ADAM10 and Bcl-w), proliferation, anchorage-independent growth, motility and migration/invasion in vitro. Intratumoral injection of the pre-miR-122-expressing BV attenuated the HCC growth/metastasis. The miR-151 sponges-expressing BV diminished the miR-151 levels for 6 weeks, enhanced RhoGDIA expression, suppressed RhoGTPases, as well as motility and migration/invasion of Mahlavu cells. Intratumoral injection of the miR-151 sponge-expressing BV impeded not only HCC metastasis but also cell proliferation, MMP expression and tumor growth in vivo. The BV co-expressing pre-miR-122 and miR-151 sponges also simultaneously enhanced miR-122 expression and inhibited miR-151, and conferred antitumor/anti-metastasis effects albeit lack of synergism. These data implicate the potentials of the SB-based hybrid BV for persistently modulating miRNA and suppressing HCC tumorigenicity/metastasis.


Subject(s)
Baculoviridae/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins pp60(c-src)/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Injections, Intralesional , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , MicroRNAs/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism
12.
Nat Protoc ; 9(8): 1882-99, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25010908

ABSTRACT

Baculovirus is a promising vector for transducing numerous types of mammalian cells. We have developed hybrid baculovirus vectors and protocols for the efficient transduction of a variety of cell lines, primary cells and stem cells, including bone marrow-derived mesenchymal stem cells (BMSCs) and adipose-derived stem cells (ASCs). The hybrid vector enables intracellular minicircle formation and prolongs transgene expression. The advantages of this transduction protocol are that baculovirus supernatant alone needs to be added to cells growing in medium, and transduction occurs after only 4-6 h of incubation at room temperature (25 °C) with gentle shaking. The entire procedure, from virus generation to transduction, can be completed within 4 weeks. Compared with other transduction procedures, this protocol is simple and can confer efficiencies >95% for many cell types. This protocol has potential applications in tissue regeneration, as transduced cells continue to express transgenes after implantation. For example, transduction of rabbit ASCs (rASCs) with growth factor-encoding hybrid baculovirus vectors, as described as an example application in this protocol, enables robust and sustained growth factor expression, stimulates stem cell differentiation and augments tissue regeneration after implantation.


Subject(s)
Baculoviridae/genetics , Transduction, Genetic/methods , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Guided Tissue Regeneration , Male , Rabbits , Stem Cells/cytology
13.
Adv Healthc Mater ; 3(9): 1486-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24652749

ABSTRACT

Graphene oxide (GO) is a nanomaterial with burgeoning bioapplications, while autophagy is implicated in cancer therapy. Although induction of autophagy by nanomaterials is reported, the underlying signaling mechanism in cancer cells and how this implicates the potential of GO in cancer therapy remain obscure. Here, it is shown that GO itself can induce the toll-like receptors (TLRs) responses and autophagy in cancer cells and confer antitumor effects in mice. GO can be phagocytosed by CT26 colon cancer cells, simultaneously triggering autophagy as well as TLR-4 and TLR-9 signaling cascades. By dissecting the crosstalk between the TLRs and autophagy pathways, it is uncovered that the GO-activated autophagy is regulated through the myeloid differentiation primary response gene 88 (MyD88)- and TNF receptor-associated factor 6 (TRAF6)-associated TLR-4/9 signaling pathways. Injection of GO alone into immunocompetent mice bearing the CT26 colon tumors not only suppresses the tumor progression but also enhances cell death, autophagy, and immune responses within the tumor bed. These data altogether implicate the potential of GO as an effective nanomaterial for autophagy induction and cancer therapy.


Subject(s)
Autophagy/drug effects , Graphite/pharmacology , Nanostructures/chemistry , Oxides/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Agents , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Graphite/chemistry , Graphite/metabolism , Mice , Oxides/chemistry , Oxides/metabolism , Phagocytosis , Toll-Like Receptors , Xenograft Model Antitumor Assays
14.
Biomaterials ; 35(18): 4901-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24674465

ABSTRACT

Repair of large calvarial bony defect remains a challenge for orthopedic surgeons. Since microRNAs (miRNAs) modulate the osteogenesis of osteoprogenitor cells, we aimed to engineer human adipose-derived stem cells (hASCs), a promising cell source for bone engineering, with miRNA-expressing baculovirus vectors. We constructed 4 baculoviruses each expressing 1 human miRNA (miR-26a, miR-29b, miR-148b, miR-196a) and verified that the miRNA-expressing baculovirus vectors augmented hASCs osteogenesis. Among these 4 miRNAs, miR-148b and miR-196a exerted more potent osteoinductive effects than miR-26a and miR-29b. Furthermore, we unveiled that co-transduction of hASCs with miR-148b-expressing and bone morphogenetic protein 2 (BMP-2)-expressing baculovirus vectors enhanced and prolonged BMP-2 expression, and synergistically promoted the in vitro osteogenic differentiation of hASCs. Implantation of the hASCs co-expressing BMP-2/miR-148b into critical-size (4 mm in diameter) calvarial bone defects in nude mice accelerated and potentiated the bone healing and remodeling, filling ≈94% of defect area and ≈89% of defect volume with native calvaria-like flat bone in 12 weeks, as judged from micro computed tomography, histology and immunohistochemical staining. Altogether, this study confirmed the feasibility of combining miRNA and growth factor expression for synergistic stimulation of in vitro osteogenesis and in vivo calvarial bone healing.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , MicroRNAs/metabolism , Osteogenesis , Skull/injuries , Stem Cells/cytology , Adipose Tissue/cytology , Animals , Baculoviridae/genetics , Bone Morphogenetic Protein 2/genetics , Cells, Cultured , Female , Gene Expression , Gene Expression Regulation , Genetic Vectors/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Tissue Engineering , Tissue Scaffolds , Wound Healing , X-Ray Microtomography
15.
Tissue Eng Part A ; 20(9-10): 1392-402, 2014 May.
Article in English | MEDLINE | ID: mdl-24367947

ABSTRACT

We previously showed that transplantation of adipose-derived stem cells (ASCs) engineered with hybrid baculovirus (BV) persistently expressing bone morphogenetic protein 2 (BMP2)/vascular endothelial growth factor (VEGF) into segmental defects in New Zealand White (NZW) rabbits led to successful defect reunion. By using microcomputed tomography and histology, here we further demonstrated that transplanting the hybrid BV-engineered ASCs into the massive defects (10 mm in length) at the femoral diaphysis of NZW rabbits resulted in trabecular bone formation in the interior via endochondral ossification and bone remodeling at 3 months post-transplantation. The progression of bone remodeling gave rise to the resorption of trabecular bone and conspicuous reconstruction of medullary cavity and cortical bone with lamellar structure at 8 months post-transplantation, hence conferring mechanical properties that were comparable to those of nonoperated femora. Importantly, X-ray, positron emission tomography/computed tomography scans, and histopathology revealed no signs of heterotopic bone formation and tumor formation. These data altogether attested that the genetically engineered ASCs and prolonged BMP2/VEGF expression not only healed and remodeled the stringent segmental defects, but also revitalized the defects into living bone tissues that structurally and biomechanically resembled intact bones without appreciable side effects, making it one step closer to translate this technology to the clinical setting.


Subject(s)
Adipocytes/cytology , Bone Morphogenetic Protein 2/therapeutic use , Femoral Fractures/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Remodeling/physiology , Cells, Cultured , Combined Modality Therapy , Femoral Fractures/diagnosis , Fracture Healing/physiology , Genetic Therapy/methods , Longitudinal Studies , Protein Engineering/methods , Rabbits , Transfection/methods , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Biomaterials ; 34(37): 9401-12, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24016854

ABSTRACT

Calvarial bone healing is difficult and grafts comprising adipose-derived stem cells (ASCs) and PLGA (poly(lactic-co-glycolic acid)) scaffolds barely heal rabbit calvarial defects. Although calvarial bone forms via intramembranous ossification without cartilage templates, it was suggested that chondrocytes/cartilages promote calvarial healing, thus we hypothesized that inducing ASCs chondrogenesis and endochondral ossification involving cartilage formation can improve calvarial healing. To evaluate this hypothesis and selectively induce osteogenesis/chondrogenesis, rabbit ASCs were engineered to express the potent osteogenic (BMP2) or chondrogenic (TGF-ß3) factor, seeded into either apatite-coated PLGA or gelatin sponge scaffolds, and allotransplanted into critical-size calvarial defects. Among the 4 ASCs/scaffold constructs, gelatin constructs elicited in vitro chondrogenesis, in vivo osteogenic metabolism and calvarial healing more effectively than apatite-coated PLGA, regardless of BMP2 or TGF-ß3 expression. The BMP2-expressing ASCs/gelatin triggered better bone healing than TGF-ß3-expressing ASCs/gelatin, filling ≈ 86% of the defect area and ≈ 61% of the volume at week 12. The healing proceeded via endochondral ossification, instead of intramembranous pathway, as evidenced by the formation of cartilage that underwent osteogenesis and hypertrophy. These data demonstrated ossification pathway switching and significantly augmented calvarial healing by the BMP2-expressing ASCs/gelatin constructs, and underscored the importance of growth factor/scaffold combinations on the healing efficacy and pathway.


Subject(s)
Adipose Tissue/cytology , Bone Morphogenetic Protein 2/genetics , Skull/injuries , Stem Cells/cytology , Tissue Scaffolds/chemistry , Transforming Growth Factor beta3/genetics , Wound Healing , Adipose Tissue/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Engineering , Cells, Cultured , Chondrogenesis , Gelatin/chemistry , Gene Expression , Lactic Acid/chemistry , Osteogenesis , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits , Stem Cells/metabolism , Transforming Growth Factor beta3/metabolism
17.
Biotechnol Adv ; 31(8): 1695-706, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994567

ABSTRACT

Gene therapy has converged with bone engineering over the past decade, by which a variety of therapeutic genes have been delivered to stimulate bone repair. These genes can be administered via in vivo or ex vivo approach using either viral or nonviral vectors. This article reviews the fundamental aspects and recent progresses in the gene therapy-based bone engineering, with emphasis on the new genes, viral vectors and gene delivery approaches.


Subject(s)
Bone and Bones , Gene Transfer Techniques , Genetic Therapy , Regenerative Medicine , Tissue Engineering , Animals , Genetic Vectors , Humans , Mice , Viruses
18.
Nucleic Acids Res ; 41(14): e139, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716635

ABSTRACT

Baculovirus (BV) is a promising gene vector but mediates transient expression. To prolong the expression, we developed a binary system whereby the transgene in the substrate BV was excised by the recombinase (ΦC31o, Cre or FLPo) expressed by a second BV and recombined into smaller minicircle. The recombination efficiency was lower by ΦC31o (≈40-75%), but approached ≈90-95% by Cre and FLPo in various cell lines and stem cells [e.g. human adipose-derived stem cells (hASCs)]. Compared with FLPo, Cre exerted higher expression level and lower negative effects; thus, we incorporated additional cis-acting element [oriP/Epstein-Barr virus nuclear antigen 1 (EBNA1), scaffold/matrix attached region or human origin of replication (ori)] into the Cre-based BV system. In proliferating cells, only oriP/EBNA1 prolonged the transgene expression and maintained the episomal minicircles for 30 days without inadvertent integration, whereas BV genome was degraded in 10 days. When delivering bmp2 or vegf genes, the efficient recombination/minicircle formation prolonged and enhanced the growth factor expression in hASCs. The prolonged bone morphogenetic protein 2 expression ameliorated the osteogenesis of hASCs, a stem cell with poor osteogenesis potential. Altogether, this BV vector exploiting Cre-mediated recombination and oriP/EBNA1 conferred remarkably high recombination efficiency, which prolonged and enhanced the transgene expression in dividing and non-dividing cells, thereby broadening the applications of BV.


Subject(s)
Baculoviridae/genetics , Recombinases/metabolism , Transgenes , Adipose Tissue/cytology , Animals , Cell Line , Cell Survival , Cells, Cultured , Cricetinae , DNA, Circular/metabolism , Gene Expression , Genetic Vectors , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Attachment Regions , Osteogenesis , Rabbits , Recombination, Genetic , Replication Origin , Stem Cells/cytology , Stem Cells/metabolism
19.
Biomaterials ; 33(30): 7422-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22796166

ABSTRACT

Baculovirus holds promise for genetic modification of adipose-derived stem cells (ASCs) and bone engineering. To explore the immune responses during bone healing and the cell fate, ASCs were mock-transduced (Mock group), transduced with the baculovirus transiently expressing growth factors promoting osteogenesis (BMP2) or angiogenesis (VEGF) (S group), or transduced with hybrid baculoviruses persistently expressing BMP2/VEGF (L group). After allotransplantation into massive femoral defects in rabbits, these 3 groups triggered similar degrees of transient inflammatory response (e.g. neutrophil proliferation and immune cell infiltration into the graft site), revealing that baculovirus and transgene products did not exacerbate the inflammation. The cells in all 3 groups underwent apoptosis initially, persisted for at least 4 weeks and were eradicated thereafter. The L group prolonged the in vivo BMP2/VEGF expression (up to 4 weeks), extended the antibody responses, and slightly enhanced the cell-mediated cytotoxicity. Nonetheless, the L group led to remarkably better bone healing and remodeling than the Mock and S groups. These data confirmed that the ASCs engineered with the hybrid BV imparted prolonged expression of BMP2/VEGF which, although stimulated low levels of humoral and cell-mediated immune responses, essentially augmented the healing of massive segmental bone defects.


Subject(s)
Adipose Tissue/cytology , Femur/immunology , Femur/pathology , Genetic Engineering , Stem Cell Transplantation , Stem Cells/immunology , Wound Healing/immunology , Animals , Antibodies/immunology , Apoptosis , Baculoviridae , Bone Morphogenetic Protein 2/metabolism , Bone Remodeling , Cell Movement , Cytotoxicity, Immunologic , Femur/diagnostic imaging , Humans , In Situ Hybridization, Fluorescence , Inflammation/pathology , Interferon-gamma/metabolism , Macrophages/cytology , Male , Rabbits , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/cytology , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Transgenes/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...