Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 28(16): 2686-2695, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31179490

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating X-linked disease affecting ~1 in 5000 males. DMD patients exhibit progressive muscle degeneration and weakness, leading to loss of ambulation and premature death from cardiopulmonary failure. We previously reported that mouse Laminin-111 (msLam-111) protein could reduce muscle pathology and improve muscle function in the mdx mouse model for DMD. In this study, we examined the ability of msLam-111 to prevent muscle disease progression in the golden retriever muscular dystrophy (GRMD) dog model of DMD. The msLam-111 protein was injected into the cranial tibial muscle compartment of GRMD dogs and muscle strength and pathology were assessed. The results showed that msLam-111 treatment increased muscle fiber regeneration and repair with improved muscle strength and reduced muscle fibrosis in the GRMD model. Together, these findings support the idea that Laminin-111 could serve as a novel protein therapy for the treatment of DMD.


Subject(s)
Laminin/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Recombinant Proteins/pharmacology , Regeneration/drug effects , Animals , Biomarkers , Disease Models, Animal , Dogs , Laminin/administration & dosage , Male , Mice , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/etiology , Phenotype , Recombinant Proteins/administration & dosage , Treatment Outcome
2.
J Am Soc Nephrol ; 29(5): 1426-1436, 2018 05.
Article in English | MEDLINE | ID: mdl-29472414

ABSTRACT

Background Laminin α5ß2γ1 (LM-521) is a major component of the GBM. Mutations in LAMB2 that prevent LM-521 synthesis and/or secretion cause Pierson syndrome, a rare congenital nephrotic syndrome with diffuse mesangial sclerosis and ocular and neurologic defects. Because the GBM is uniquely accessible to plasma, which permeates endothelial cell fenestrae, we hypothesized that intravenous delivery of LM-521 could replace the missing LM-521 in the GBM of Lamb2 mutant mice and restore glomerular permselectivity.Methods We injected human LM-521 (hLM-521), a macromolecule of approximately 800 kD, into the retro-orbital sinus of Lamb2-/- pups daily. Deposition of hLM-521 into the GBM was investigated by fluorescence microscopy. We assayed the effects of hLM-521 on glomerular permselectivity by urinalysis and the effects on podocytes by desmin immunostaining and ultrastructural analysis of podocyte architecture.Results Injected hLM-521 rapidly and stably accumulated in the GBM of all glomeruli. Super-resolution imaging showed that hLM-521 accumulated in the correct orientation in the GBM, primarily on the endothelial aspect. Treatment with hLM-521 greatly reduced the expression of the podocyte injury marker desmin and attenuated the foot process effacement observed in untreated pups. Moreover, treatment with hLM-521 delayed the onset of proteinuria but did not prevent nephrotic syndrome, perhaps due to its absence from the podocyte aspect of the GBM.Conclusions These studies show that GBM composition and function can be altered in vivovia vascular delivery of even very large proteins, which may advance therapeutic options for patients with abnormal GBM composition, whether genetic or acquired.


Subject(s)
Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/metabolism , Eye Abnormalities/drug therapy , Eye Abnormalities/metabolism , Glomerular Basement Membrane/metabolism , Laminin/genetics , Laminin/therapeutic use , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/metabolism , Pupil Disorders/drug therapy , Pupil Disorders/metabolism , Abnormalities, Multiple/genetics , Animals , Desmin/metabolism , Disease Models, Animal , Eye Abnormalities/complications , Eye Abnormalities/genetics , Injections, Intravenous , Laminin/administration & dosage , Mice , Myasthenic Syndromes, Congenital , Nephrotic Syndrome/complications , Nephrotic Syndrome/etiology , Nephrotic Syndrome/genetics , Permeability/drug effects , Podocytes/drug effects , Podocytes/metabolism , Podocytes/ultrastructure , Proteinuria/etiology , Proteinuria/prevention & control , Pupil Disorders/complications , Pupil Disorders/genetics , Recombinant Proteins/therapeutic use
3.
Biol Res ; 51(1): 15, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29866159

ABSTRACT

BACKGROUND: Translational research to develop pharmaceutical and surgical treatments for pterygium requires a reliable and easy to produce animal model. Extracellular matrix and fibroblast are important components of pterygium. The aim of this study was to analyze the effect of the subconjunctival injection of fibroblast cells (NIH3T3 cell line) and exogenous extracellular matrix in rabbits in producing a pterygium-like lesion. METHODS: Six 3-month-old white New Zealand rabbits were injected with 20,000 NIH3T3 cells and 5 µL of Matrigel in the right conjunctiva, and with only 5 µL of Matrigel in the left conjunctiva. The eyes were photographed under a magnification of 16× using a 12-megapixel digital camera attached to the microscope on day 1, 3 and 7. Conjunctival vascularization was measured by analyzing images to measure red pixel saturation. Area of corneal and conjunctival fibrovascular tissue formation on the site of injection was assessed by analyzing the images on day 3 and 7 using area measurement software. Histopathologic characteristics were determined in the rabbit tissues and compared with a human primary pterygium. RESULTS: The two treatments promoted growth of conjunctival fibrovascular tissue at day 7. The red pixel saturation and area of fibrovascular tissue developed was significantly higher in right eyes (p < 0.05). Tissues from both treatments showed neovascularization in lesser extent to that observed in human pterygium. Acanthosis, stromal inflammation, and edema were found in tissues of both treatments. No elastosis was found in either treatment. CONCLUSIONS: Matrigel alone or in combination with NIH3T3 cells injected into the rabbits' conjunctiva can promote tissue growth with characteristics of human pterygium, including neovascularization, acanthosis, stromal inflammation, and edema. The combination of Matrigel with NIH3T3 cells seems to have an additive effect on the size and redness of the pterygium-like tissue developed.


Subject(s)
Collagen/adverse effects , Disease Models, Animal , Extracellular Matrix/transplantation , Fibroblasts/transplantation , Laminin/adverse effects , Proteoglycans/adverse effects , Pterygium/etiology , Animals , Collagen/administration & dosage , Drug Combinations , Laminin/administration & dosage , Mice , NIH 3T3 Cells , Proteoglycans/administration & dosage , Pterygium/pathology , Rabbits
4.
Circ Res ; 117(12): 995-1000, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26429802

ABSTRACT

RATIONALE: The lack of measurable single-cell contractility of human-induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs) currently limits the utility of hiPSC-CMs for evaluating contractile performance for both basic research and drug discovery. OBJECTIVE: To develop a culture method that rapidly generates contracting single hiPSC-CMs and allows quantification of cell shortening with standard equipment used for studying adult CMs. METHODS AND RESULTS: Single hiPSC-CMs were cultured for 5 to 7 days on a 0.4- to 0.8-mm thick mattress of undiluted Matrigel (mattress hiPSC-CMs) and compared with hiPSC-CMs maintained on a control substrate (<0.1-mm thick 1:60 diluted Matrigel, control hiPSC-CMs). Compared with control hiPSC-CMs, mattress hiPSC-CMs had more rod-shape morphology and significantly increased sarcomere length. Contractile parameters of mattress hiPSC-CMs measured with video-based edge detection were comparable with those of freshly isolated adult rabbit ventricular CMs. Morphological and contractile properties of mattress hiPSC-CMs were consistent across cryopreserved hiPSC-CMs generated independently at another institution. Unlike control hiPSC-CMs, mattress hiPSC-CMs display robust contractile responses to positive inotropic agents, such as myofilament calcium sensitizers. Mattress hiPSC-CMs exhibit molecular changes that include increased expression of the maturation marker cardiac troponin I and significantly increased action potential upstroke velocity because of a 2-fold increase in sodium current (INa). CONCLUSIONS: The Matrigel mattress method enables the rapid generation of robustly contracting hiPSC-CMs and enhances maturation. This new method allows quantification of contractile performance at the single-cell level, which should be valuable to disease modeling, drug discovery, and preclinical cardiotoxicity testing.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Cell Differentiation/drug effects , Cells, Cultured , Collagen/administration & dosage , Drug Combinations , Humans , Induced Pluripotent Stem Cells/drug effects , Laminin/administration & dosage , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Proteoglycans/administration & dosage
5.
Biomacromolecules ; 18(9): 2723-2731, 2017 Sep 11.
Article in English | MEDLINE | ID: mdl-28813139

ABSTRACT

A PEG-based cyclized vinyl polymer was synthesized via one-step RAFT polymerization and used as a precursor of injectable hydrogels. Dithiol linkers including laminin-derived peptides containing IKVAV and YIGSR sequences and DTT were used for gelation. Fast and adjustable gelation rate was achieved through nucleophile-initiated thiol-Michael reaction under physiological conditions. Low swelling ratio and moderate degradation rate of the formed hydrogels were observed. 3D encapsulation of neural progenitor cells in the synthetic hydrogel showed good cell viability over 8 days. The long-term cell survival and proliferation were promoted by the introduction of laminin-derived peptides. This hydrogel platform based on peptide-cross-linked, cyclized vinyl polymers can be used as a universal hydrogel template for 3D cell encapsulation.


Subject(s)
Hydrogels/chemistry , Laminin/chemistry , Nanoparticles/chemistry , Oligopeptides/chemistry , Peptide Fragments/chemistry , Vinyl Compounds/chemistry , Cell Proliferation , Cell Survival , HeLa Cells , Humans , Hydrogels/adverse effects , Hydrogels/chemical synthesis , Laminin/administration & dosage , Nanoparticles/adverse effects , Neural Stem Cells/drug effects , Oligopeptides/administration & dosage , Peptide Fragments/administration & dosage , Vinyl Compounds/adverse effects , Vinyl Compounds/chemical synthesis
6.
Hum Mol Genet ; 23(2): 383-96, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24009313

ABSTRACT

Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a severe and fatal muscle-wasting disease with no cure. MDC1A patients and the dy(W-/-) mouse model exhibit severe muscle weakness, demyelinating neuropathy, failed muscle regeneration and premature death. We have recently shown that laminin-111, a form of laminin found in embryonic skeletal muscle, can substitute for the loss of laminin-211/221 and prevent muscle disease progression in the dy(W-/-) mouse model. What is unclear from these studies is whether laminin-111 can restore failed regeneration to laminin-α2-deficient muscle. To investigate the potential of laminin-111 protein therapy to improve muscle regeneration, laminin-111 or phosphate-buffered saline-treated laminin-α2-deficient muscle was damaged with cardiotoxin and muscle regeneration quantified. Our results show laminin-111 treatment promoted an increase in myofiber size and number, and an increased expression of α7ß1 integrin, Pax7, myogenin and embryonic myosin heavy chain, indicating a restoration of the muscle regenerative program. Together, our results show laminin-111 restores muscle regeneration to laminin-α2-deficient muscle and further supports laminin-111 protein as a therapy for the treatment of MDC1A.


Subject(s)
Laminin/pharmacology , Muscular Atrophy/drug therapy , Muscular Dystrophies/drug therapy , Muscular Dystrophies/pathology , Animals , Fibrosis/drug therapy , Gene Expression Regulation , Laminin/administration & dosage , Mice , Muscular Atrophy/metabolism , Muscular Dystrophy, Animal , Myogenin/metabolism , PAX7 Transcription Factor/metabolism
7.
Exp Eye Res ; 143: 60-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26474493

ABSTRACT

Nanofiber-based hydrogels (nanogels) with different, covalently bound peptides were used as an extracellular environment for lens epithelial cells (LECs) in order to modulate the capsular opacification (CO) response after lens surgery in a porcine eye model. Lenses were divided into 15 groups (n = 4 per group), the lens content was removed and the empty capsules were refilled with nanogel without peptides and nanogels with 13 combinations of 5 different peptides: two laminin-derived, two fibronectin-derived, and one collagen IV-derived peptide representing cell adhesion motifs. A control group of 4 lenses was refilled with hyaluronan. After refilling, lenses were extracted from the porcine eye and cultured for three weeks. LECs were assessed for morphology and alpha smooth muscle actin (αSMA) expression using confocal laser scanning microscopy. Compared to hyaluronan controls, lenses filled with nanogel had less CO formation, indicated by a lower αSMA expression (P = 0.004). Microscopy showed differences in morphological cell response within the nanogel refilled groups. αSMA expression in these groups was highest in lenses refilled with nanogel without peptides (9.54 ± 11.29%). Overall, LEC transformation is reduced by the presence of nanogels and the response is improved even further by incorporation of extracellular matrix peptides representing adhesion motifs. Thus, nanomaterials targeting biological pathways, in our case interactions with integrin signaling, are a promising avenue toward reduction of CO. Further research is needed to optimize nanogel-peptide combinations that fully prevent CO.


Subject(s)
Capsule Opacification/prevention & control , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix Proteins/administration & dosage , Hydrogels , Lens Capsule, Crystalline/cytology , Oligopeptides/administration & dosage , Actins/metabolism , Animals , Biomarkers/metabolism , Capsule Opacification/pathology , Collagen Type IV/administration & dosage , Collagen Type IV/chemical synthesis , Drug Delivery Systems , Extracellular Matrix Proteins/chemical synthesis , Fibronectins/administration & dosage , Fibronectins/chemical synthesis , Fluorescent Antibody Technique, Indirect , Laminin/administration & dosage , Laminin/chemical synthesis , Lens, Crystalline/cytology , Nanofibers , Oligopeptides/chemical synthesis , Organ Culture Techniques , Sus scrofa
8.
J Vasc Res ; 52(2): 116-26, 2015.
Article in English | MEDLINE | ID: mdl-26277802

ABSTRACT

BACKGROUND: Angiogenesis, the formation of new blood vessels, is an essential process under physiological and pathological conditions. METHOD: Here, we improved the directed in vivo angiogenesis assay (DIVAA®) test, which is based on the usage of small Matrigel-filled tubes that are implanted into mice subcutaneously for a period of up to 15 days. The subsequent ex vivo assessment of neoangiogenesis within the silicon tubes is then achieved by fluorometry. RESULTS: We showed that the immunohistochemical quantification of the ingrowth of endothelial cells, based on CD31, was superior to the fluorometric quantification advised in the manufacturer's instructions. We optimised the explantation procedure, ensuring the complete recovery of the ingrown vessels. Using this modified protocol, we investigated the effect of the length of stay of the implanted tubes as well as of the concentration of the growth factors VEGF and FGF on the assay. CONCLUSION: Our improved protocol offered an effective and reliable alternative to the original assay, which is expected to facilitate in vivo research on angiogenesis and, thus, might drive the development of novel therapeutic agents.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Biological Assay/methods , Collagen/administration & dosage , Endothelial Cells/drug effects , Immunohistochemistry , Laminin/administration & dosage , Neovascularization, Physiologic/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proteoglycans/administration & dosage , Subcutaneous Tissue/blood supply , Animals , Biomarkers/metabolism , Drug Combinations , Endothelial Cells/metabolism , Fluorometry , Image Processing, Computer-Assisted , Mice, Inbred C57BL , Reproducibility of Results , Time Factors , Vascular Endothelial Growth Factor A/administration & dosage
9.
Pharm Res ; 31(9): 2453-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24633416

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of locally delivered pancreatic islet with liposomal clodronate (Clodrosome®) as an immunoprotection agent for the treatment of type 1 diabetes. METHOD: The bio-distribution of liposomal clodronate in matrigel was checked by imaging analyzer. To verify the therapeutic efficacy of locally delivered islet with liposomal clodronate using injectable hydrogel, four groups of islet transplanted mice (n = 6 in each group) were prepared: 1) the islet group, 2) the islet-Clodrosome group, 3) the islet-Matrigel group, and 4) the islet-Matrigel-Clodrosome group. Immune cell migration and activation, and pro-inflammatory cytokine secretion was evaluated by immunohistochemistry staining and ELISA assay. RESULTS: Cy5.5 labeled liposomes remained in the matrigel for over 7 days. The median survival time of transplanted islets (Islet-Matrigel-Clodrosome group) was significantly increased (>60 days), compared to other groups. Locally delivered liposomal clodronate in matrigel effectively inhibited the activation of macrophages, immune cell migration and activation, and pro-inflammatory cytokine secretion from macrophages. CONCLUSIONS: Locally co-delivered pancreatic islets and liposomal clodronate using injectable hydrogel effectively cured type 1 diabetes. Especially, the inhibition of macrophage attack in the early stage after local delivery of islets was very important for the successful long-term survival of delivered islets.


Subject(s)
Clodronic Acid/administration & dosage , Collagen/administration & dosage , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Laminin/administration & dosage , Proteoglycans/administration & dosage , Animals , Clodronic Acid/therapeutic use , Collagen/therapeutic use , Diabetes Mellitus, Type 1/immunology , Drug Combinations , Inflammation/immunology , Inflammation/prevention & control , Injections , Laminin/therapeutic use , Liposomes , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Proteoglycans/therapeutic use , Rats, Sprague-Dawley
10.
Angiogenesis ; 16(4): 847-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23775497

ABSTRACT

Prostate specific membrane antigen (PSMA) is a pro-angiogenic cell-surface protease that we previously demonstrated regulates blood vessel formation in a laminin and integrin ß1-dependent manner. Here, we examine the principal mechanism of PSMA activation of integrin ß1. We show that digesting laminin sequentially with recombinant matrix metalloprotease-2 (MMP-2) and PSMA generates small peptides that enhance endothelial cell adhesion and migration in vitro. We also provide evidence that these laminin peptides activate adhesion via integrin α6ß1 and focal adhesion kinase. Using an in vivo Matrigel implant assay, we show that these MMP/PSMA-derived laminin peptides also increase angiogenesis in vivo. Together, our results reveal a novel mechanism of PSMA activation of angiogenesis by processing laminin downstream of MMP-2.


Subject(s)
Antigens, Surface/physiology , Glutamate Carboxypeptidase II/physiology , Laminin/metabolism , Matrix Metalloproteinase 2/metabolism , Neovascularization, Physiologic/drug effects , Animals , Cell Adhesion , Cell Movement , Collagen/metabolism , Drug Combinations , Drug Implants , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , Human Umbilical Vein Endothelial Cells , Integrin alpha6beta1/physiology , Laminin/administration & dosage , Laminin/pharmacology , Mice , Mice, Inbred C57BL , Microvessels/growth & development , Peptide Fragments/administration & dosage , Peptide Fragments/biosynthesis , Peptide Fragments/pharmacology , Protein Processing, Post-Translational , Proteoglycans , Recombinant Proteins/metabolism , Substrate Specificity
11.
Am J Pathol ; 180(4): 1593-602, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22322301

ABSTRACT

Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a lethal muscle-wasting disease that is caused by mutations in the LAMA2 gene, resulting in the loss of laminin-α2 protein. MDC1A patients exhibit severe muscle weakness from birth, are confined to a wheelchair, require ventilator assistance, and have reduced life expectancy. There are currently no effective treatments or cures for MDC1A. Laminin-α2 is required for the formation of heterotrimeric laminin-211 (ie, α2, ß1, and γ1) and laminin-221 (ie, α2, ß2, and γ1), which are major constituents of skeletal muscle basal lamina. Laminin-111 (ie, α1, ß1, and γ1) is the predominant laminin isoform in embryonic skeletal muscle and supports normal skeletal muscle development in laminin-α2-deficient muscle but is absent from adult skeletal muscle. In this study, we determined whether treatment with Engelbreth-Holm-Swarm-derived mouse laminin-111 protein could rescue MDC1A in the dy(W-/-) mouse model. We demonstrate that laminin-111 protein systemically delivered to the muscles of laminin-α2-deficient mice prevents muscle pathology, improves muscle strength, and dramatically increases life expectancy. Laminin-111 also prevented apoptosis in laminin-α2-deficient mouse muscle and primary human MDC1A myogenic cells, which indicates a conserved mechanism of action and cross-reactivity between species. Our results demonstrate that laminin-111 can serve as an effective protein substitution therapy for the treatment of muscular dystrophy in the dy(W-/-) mouse model and establish the potential for its use in the treatment of MDC1A.


Subject(s)
Laminin/therapeutic use , Muscular Dystrophies/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Fibrosis , Humans , Injections, Intramuscular , Injections, Intraperitoneal , Kaplan-Meier Estimate , Laminin/administration & dosage , Laminin/deficiency , Laminin/metabolism , Mice , Motor Activity/drug effects , Muscle Strength/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Myoblasts/drug effects , Myoblasts/pathology , Myositis/prevention & control , Protein Isoforms/administration & dosage , Protein Isoforms/therapeutic use , Weight Loss/drug effects
12.
Nanomedicine ; 9(6): 795-805, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23434679

ABSTRACT

Matrigel, a mouse sarcoma-derived basement membrane protein mixture, is frequently used to facilitate human tumor xenograft growth in rodents. Despite its known effects on tumor growth and metastasis, its impact on tumor pathophysiology and preclinical evaluation of nanomedicines in tumor xenografts has not been reported previously. Herein bilateral MDA435 tumors were established orthotopically with (Mat+) or without (Mat-) co-injection of Matrigel. Tumor perfusion, morphology and nanoparticle retention were evaluated. As compared to Mat- tumors, Mat+tumors exhibited enhanced vascular perfusion and lymphatic flow, greater blood vessel and lymphatic growth within the tumor core, and more deformation and collapse of lymphatics in tumor-associated lymph nodes. These changes were accompanied by reduced nanoparticle retention in Mat+tumors. The results suggest that Matrigel is not a passive medium for tumor growth, but rather significantly alters long-term tumor architecture. These findings have significant implications for the evaluation of therapeutic nanomedicine in xenograft mouse models. FROM THE CLINICAL EDITOR: Matrigel is utilized in facilitating human tumor xenograft growth in rodents. The authors demonstrate that Matrigel is not a passive medium for tumor growth; instead it significantly alters long-term tumor architecture, with major implications in the evaluation of therapeutic nanomedicine in xenograft mouse models.


Subject(s)
Adenocarcinoma/physiopathology , Breast Neoplasms/physiopathology , Collagen/administration & dosage , Heterografts/physiopathology , Laminin/administration & dosage , Proteoglycans/administration & dosage , Animals , Cell Line, Tumor , Collagen/metabolism , Drug Combinations , Female , Humans , Laminin/metabolism , Mice , Nanomedicine , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Proteoglycans/metabolism
13.
J Mammary Gland Biol Neoplasia ; 17(2): 99-101, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22581302

ABSTRACT

The analysis of normal mammary morphogenesis is facilitated by the use of mammary fat pad transplantation. The recent experiments on analysis of normal mammary epithelial stem cell activity rely heavily on this technique. In this review, we discuss the known and unknown attributes of using Matrigel in the injection of the mammary epithelial cell suspension. Matrigel greatly increases the "take" frequency of the injected cell suspension; however, there is some uncertainty regarding the interpretation of some of the results. After consideration of these issues, our conclusion is that Matrigel is important in order to obtain rigorous and reproducible results.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/transplantation , Biocompatible Materials/administration & dosage , Collagen/administration & dosage , Laminin/administration & dosage , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Models, Biological , Morphogenesis , Proteoglycans/administration & dosage , Adult Stem Cells/metabolism , Animals , Biocompatible Materials/metabolism , Cell Differentiation , Cell Growth Processes , Cells, Cultured , Collagen/metabolism , Drug Combinations , Epithelium/metabolism , Female , Graft Survival , Humans , Laminin/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Proteoglycans/metabolism , Sarcoma/metabolism , Stem Cell Niche
14.
J Cell Biochem ; 113(11): 3380-92, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22688921

ABSTRACT

Osteosarcoma is the second leading cause of cancer-related death for children and young adults. In this study, we have subcutaneously injected-with and without matrigel-athymic mice (Fox1nu/nu) with human osteosarcoma 3AB-OS pluripotent cancer stem cells (CSCs), which we previously isolated from human osteosarcoma MG63 cells. Engrafted 3AB-OS cells were highly tumorigenic and matrigel greatly accelerated both tumor engraftment and growth rate. 3AB-OS CSC xenografts lacked crucial regulators of beta-catenin levels (E-cadherin, APC, and GSK-3beta), and crucial factors to restrain proliferation, resulting therefore in a strong proliferation potential. During the first weeks of engraftment 3AB-OS-derived tumors expressed high levels of pAKT, beta1-integrin and pFAK, nuclear beta-catenin, c-Myc, cyclin D2, along with high levels of hyperphosphorylated-inactive pRb and anti-apoptotic proteins such as Bcl-2 and XIAP, and matrigel increased the expression of proliferative markers. Thereafter 3AB-OS tumor xenografts obtained with matrigel co-injection showed decreased proliferative potential and AKT levels, and undetectable hyperphosphorylated pRb, whereas beta1-integrin and pFAK levels still increased. Engrafted tumor cells also showed multilineage commitment with matrigel particularly favoring the mesenchymal lineage. Concomitantly, many blood vessels and muscle fibers appeared in the tumor mass. Our findings suggest that matrigel might regulate 3AB-OS cell behavior providing adequate cues for transducing proliferation and differentiation signals triggered by pAKT, beta1-integrin, and pFAK and addressed by pRb protein. Our results provide for the first time a mouse model that recapitulates in vivo crucial features of human osteosarcoma CSCs that could be used to test and predict the efficacy in vivo of novel therapeutic treatments.


Subject(s)
Bone Neoplasms/genetics , Collagen/administration & dosage , Laminin/administration & dosage , Neoplastic Stem Cells/transplantation , Osteosarcoma/genetics , Pluripotent Stem Cells/transplantation , Proteoglycans/administration & dosage , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Collagen/metabolism , Drug Combinations , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Injections, Subcutaneous , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Laminin/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Proteoglycans/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transplantation, Heterologous
15.
J Cell Mol Med ; 15(6): 1310-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20477905

ABSTRACT

Matrigel promotes angiogenesis in the myocardium from ischemic injury and prevents remodelling of the left ventricle. We assessed the therapeutic efficacy of intracardiac matrigel injection and matrigel-mediated stem cell homing in a rat myocardial infarction (MI) model. Following MI, matrigel (250 µl) or phosphate-buffered solution (PBS) was delivered by intracardiac injection. Compared to the MI control group (MI-PBS), matrigel significantly improved left ventricular function (n= 11, P < 0.05) assessed by pressure-volume loops after 4 weeks. There is no significant difference in infarct size between MI-matrigel (MI-M; 21.48 ± 1.49%, n = 10) and MI-PBS hearts (20.98 ± 1.25%, n = 10). The infarct wall thickness of left ventricle is significantly higher (P < 0.01) in MI-M (0.72 ± 0.02 mm, n = 10) compared with MI-PBS (0.62 ± 0.02 mm, n = 10). MI-M hearts exhibited higher capillary density (border 130.8 ± 4.7 versus 115.4 ± 6.0, P < 0.05; vessels per high-power field [HPF; 400×], n = 6) than MI-PBS hearts. c-Kit(+) stem cells (38.3 ± 5.3 versus 25.7 ± 1.5 c-Kit(+) cells per HPF [630×], n = 5, P < 0.05) and CD34(+) cells (13.0 ± 1.51 versus 5.6 ± 0.68 CD34(+) cells per HPF [630×], n = 5, P < 0.01) were significantly more numerous in MI-M than in MI-PBS in the infarcted hearts (n = 5, P < 0.05). Intracardiac matrigel injection restores myocardial functions following MI, which may attribute to the improved recruitment of CD34(+) and c-Kit(+) stem cells.


Subject(s)
Cell Movement/drug effects , Collagen , Laminin , Myocardial Infarction/drug therapy , Myocardium/pathology , Proteoglycans , Animals , Aorta, Thoracic/physiopathology , Collagen/administration & dosage , Collagen/therapeutic use , Disease Models, Animal , Drug Combinations , Hemodynamics/drug effects , Injections, Intramuscular , Laminin/administration & dosage , Laminin/therapeutic use , Ligation , Male , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Proteoglycans/administration & dosage , Proteoglycans/therapeutic use , Rats , Rats, Inbred Strains , Stem Cells/physiology , Ventricular Function, Left/drug effects
16.
Cancer Treat Res Commun ; 27: 100315, 2021.
Article in English | MEDLINE | ID: mdl-33571801

ABSTRACT

Models of human cancer, to be useful, must replicate human disease with high fidelity. Our focus in this study is rat xenograft brain tumors as a model of human embedded cerebral tumors. A distinguishing signature of such tumors in humans, that of contrast-enhancement on imaging, is often not present when the human cells grow in rodents, despite the xenografts having nearly identical DNA signatures to the original tumor specimen. Although contrast enhancement was uniformly evident in all the human tumors from which the xenografts' cells were derived, we show that long-term contrast enhancement in the model tumors may be determined conditionally by the tumor microenvironment at the time of cell implantation. We demonstrate this phenomenon in one of two patient-derived orthotopic xenograft (PDOX) models using cancer stem-like cell (CSC)-enriched neurospheres from human tumor resection specimens, transplanted to groups of immune-compromised rats in the presence or absence of a collagen/fibrin scaffolding matrix, Matrigel. The rats were imaged by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and their brains were examined by histopathology. Targeted proteomics of the PDOX tumor specimens grown from CSC implanted with and without Matrigel showed that while the levels of the majority of proteins and post-translational modifications were comparable between contrast-enhancing and non-enhancing tumors, phosphorylation of Fox038 showed a differential expression. The results suggest key proteins determine contrast enhancement and suggest a path toward the development of better animal models of human glioma. Future work is needed to elucidate fully the molecular determinants of contrast-enhancement.


Subject(s)
Brain Neoplasms/diagnosis , Brain/diagnostic imaging , Collagen/administration & dosage , Glioblastoma/diagnosis , Laminin/administration & dosage , Proteoglycans/administration & dosage , Tumor Microenvironment , Animals , Brain/pathology , Brain Neoplasms/pathology , Drug Combinations , Female , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Neoplastic Stem Cells/pathology , Rats , Spheroids, Cellular , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
17.
Sci Rep ; 11(1): 22243, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34782616

ABSTRACT

Although endogenous cardiac repair by recruitment of stem cells may serve as a therapeutic approach to healing a damaged heart, how to effectively enhance the migration of stem cells to the damaged heart is unclear. Here, we examined whether the combined administration of prostacyclin agonist (ONO1301), a multiple-cytokine inducer, and stem cell niche laminin-221 (LM221), enhances regeneration through endogenous cardiac repair. We administered ONO1301- and LM221-immersed sheets, LM221-immersed sheets, ONO1301-immersed sheets, and PBS-immersed sheets (control) to an acute infarction rat model. Four weeks later, cardiac function, histology, and cytokine expression were analysed. The combined administration of LM221 and ONO1301 upregulated angiogenic and chemotactic factors in the myocardium after 4 weeks and enhanced the accumulation of ILB4 positive cells, SMA positive cells, and platelet-derived growth factor receptor alpha (PDGFRα) and CD90 double-positive cells, leading to the generation of mature microvascular networks. Interstitial fibrosis reduced and functional recovery was prominent in LM221- and ONO1301-administrated hearts as compared with those in ONO1301-administrated or control hearts. LM221 and ONO1301 combination enhanced recruitment of PDGFRα and CD90 double-positive cells, maturation of vessels, and functional recovery in rat acute myocardial infarction hearts, highlighting a new promising acellular approach for the failed heart.


Subject(s)
Epoprostenol/administration & dosage , Laminin/administration & dosage , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Wound Healing/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Drug Therapy, Combination , Gene Expression Regulation/drug effects , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protective Agents/pharmacology , Rats , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Regeneration/drug effects , Thy-1 Antigens/metabolism , Treatment Outcome
18.
J Neurosci Res ; 88(14): 3161-70, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20818775

ABSTRACT

Injection into the injured spinal cord of peptide amphiphile (PA) molecules that self-assemble and display the laminin epitope IKVAV at high density improved functional recovery after spinal cord injury (SCI) in two different species, rat and mouse, and in two different injury models, contusion and compression. The improvement required the IKVAV epitope and was not observed with the injection of an amphiphile displaying a nonbioactive sequence. To explore the mechanisms underlying these improvements, the number of serotonergic fibers in the lesioned spinal cord was compared in animals receiving the IKVAV-PA, a nonbioactive PA (PA control), or sham injection. Serotonergic fibers were distributed equally in all three groups rostral to the injury but showed a significantly higher density caudal to the injury site in the IKVAV PA-injected group. Furthermore, this difference was not present in the subacute phase following injury but appeared in the chronically injured cord. The IKVAV PA-injected groups also trended higher both in the total number neurons adjacent to the lesion and in the number of long propriospinal tract connections from the thoracic to the lumbar cord. IKVAV PA injection did not alter myelin thickness, total axon number caudal to the lesion, axon size distribution, or total axon area. Serotonin can promote stepping even in complete transection models, so the improved function produced by the IKVAV PA treatment may reflect the increased serotonergic innervation caudal to the lesion in addition to the previously demonstrated regeneration of motor and sensory axons through the lesion.


Subject(s)
Nerve Fibers/drug effects , Neuronal Plasticity/drug effects , Serotonin/physiology , Spinal Cord Injuries/drug therapy , Surface-Active Agents/pharmacology , Animals , Cell Count , Disease Models, Animal , Female , Laminin/administration & dosage , Laminin/physiology , Mice , Mice, 129 Strain , Nanofibers , Nerve Fibers/physiology , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Peptide Fragments/administration & dosage , Peptides/administration & dosage , Rats , Rats, Long-Evans , Spinal Cord Injuries/metabolism
19.
J Autoimmun ; 35(3): 232-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20655705

ABSTRACT

Beta 2 glycoprotein I (ß2GPI) is the major auto antigen in the antiphospholipid syndrome but also interacts with fibrinolytic and angiogenic proteins. The aim of this study was to examine the angiogenic potential of ß2GPI in vivo in ß2GPI deficient mice utilizing angiogenic assays. ß2GPI deficient mice show increased microvessel formation in comparison to ß2GPI replete controls when injected with growth factor free-matrigel implants. However, microvessel formation in matrigel plugs of ß2GPI deficient mice was less than in ß2GPI replete mice when basic fibroblast growth factor (bFGF) was included in the matrigel. Hemoglobin content was higher in vascular endothelial growth factor (VEGF) containing-matrigel plugs in the ß2GPI deficient mouse demonstrating that the lack of ß2GPI led to increased extravasation by VEGF. Melanoma B16F10 tumour growth was enhanced in ß2GPI deficient mice. Melanoma microvessel density was increased in ß2GPI deficient mice but the proliferation rate of tumour cells (determined by Ki67 immunohistochemistry) was unaffected by the presence or absence of ß2GPI. Subcutaneous delivery of native human ß2GPI by the ALZET osmotic pump did not affect melanoma tumour growth in ß2GPI deficient mice. We conclude that the in vivo unopposed action of ß2GPI is anti-angiogenic however this function is modified in the presence of a strong angiogenic stimulus into stabilization of vessel formation. Although the presence of ß2GPI attenuates vessel sprouting in certain tumours, no survival benefit is conferred to tumour bearing animals. This does not preclude the potential benefit of modified or fragments of ß2GPI in anti-angiogenesis research.


Subject(s)
Melanoma, Experimental/blood supply , Microvessels/metabolism , Neovascularization, Pathologic , Recombinant Proteins/metabolism , Skin Neoplasms/blood supply , beta 2-Glycoprotein I/metabolism , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line , Collagen/administration & dosage , Drug Combinations , Fibroblast Growth Factor 2/administration & dosage , Humans , Infusion Pumps , Laminin/administration & dosage , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Microvessels/pathology , Neovascularization, Pathologic/genetics , Proteoglycans/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Burden/drug effects , Tumor Burden/genetics , Vascular Endothelial Growth Factor A/administration & dosage , beta 2-Glycoprotein I/administration & dosage , beta 2-Glycoprotein I/genetics
20.
Mol Pharm ; 7(1): 217-26, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20020739

ABSTRACT

A promising strategy as a cancer therapeutic is tumor-targeted gene delivery. The AG73 peptide derived from the laminin alpha1 chain is a ligand for syndecans, and syndecan-2 is highly expressed in some cancer cells. In this study, AG73-PEG liposomes were developed for selective gene delivery to syndecan-2 overexpressing cancer cells. AG73-PEG liposomes were used in combination with Bubble liposomes and ultrasound exposure to enhance transfection efficiency by promoting the escape of the liposomes from the endosome to the cytosol. AG73-PEG liposomes showed selective gene delivery to syndecan-2 overexpressing cancer cells. Furthermore, AG73-mediated liposomal gene transfection efficiency was enhanced by 60-fold when Bubble liposomes and ultrasound exposure were used, despite the absence of an increase in the uptake of AG73-PEG liposomes into the cells. Confocal microscope analysis revealed that the Bubble liposomes and ultrasound promoted intracellular trafficking of the AG73-PEG liposomes during gene transfection. Thus, the combination of AG73-PEG liposomes with Bubble liposomes and ultrasound exposure may be a promising method to achieve selective and efficient gene delivery for cancer therapy.


Subject(s)
Gene Transfer Techniques , Cell Line, Tumor , Drug Delivery Systems , Genetic Therapy/methods , Humans , Laminin/administration & dosage , Laminin/chemistry , Liposomes , Microbubbles , Peptides/administration & dosage , Peptides/chemistry , Plasmids/administration & dosage , Plasmids/genetics , Polyethylene Glycols , Syndecan-2/metabolism , Transfection , Ultrasonics
SELECTION OF CITATIONS
SEARCH DETAIL