Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cells ; 10(11)2021 11 08.
Article in English | MEDLINE | ID: mdl-34831300

ABSTRACT

Human cytomegalovirus (HCMV) infection is an important cause of morbidity and mortality in immunocompromised patients and a major etiological factor for congenital birth defects in newborns. Ganciclovir and its pro-drug valganciclovir are the preferred drugs in use today for prophylaxis and treatment of viremic patients. Due to long treatment times, patients are at risk for developing viral resistance to ganciclovir and to other drugs with a similar mechanism of action. We earlier found that the endothelin receptor B (ETBR) is upregulated during HCMV infection and that it plays an important role in the life cycle of this virus. Here, we tested the hypothesis that ETBR blockade could be used in the treatment of HCMV infection. As HCMV infection is specific to humans, we tested our hypothesis in human cell types that are relevant for HCMV pathogenesis; i.e., endothelial cells, epithelial cells and fibroblasts. We infected these cells with HCMV and treated them with the ETBR specific antagonist BQ788 or ETR antagonists that are approved by the FDA for treatment of pulmonary hypertension; macitentan, its metabolite ACT-132577, bosentan and ambrisentan, and as an anti-viral control, we used ganciclovir or letermovir. At concentrations expected to be relevant in vivo, macitentan, ACT-132577 and BQ788 effectively inhibited productive infection of HCMV. Of importance, macitentan also inhibited productive infection of a ganciclovir-resistant HCMV isolate. Our results suggest that binding or signaling through ETBR is crucial for viral replication, and that selected ETBR blockers inhibit HCMV infection.


Subject(s)
Cytomegalovirus/physiology , Endothelin Receptor Antagonists/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus Infections/virology , Endothelin Receptor Antagonists/chemistry , Ganciclovir/pharmacology , Humans , Inhibitory Concentration 50 , Metabolome , Oligopeptides , Piperidines , Pyrimidines/chemistry , Sulfonamides/chemistry , Virion/drug effects , Virion/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects
2.
Cell Chem Biol ; 28(12): 1693-1702.e6, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34192523

ABSTRACT

Ganciclovir (GCV) is the first-line therapy against human cytomegalovirus (HCMV), a widespread infection that is particularly dangerous for immunodeficient individuals. Closely resembling deoxyguanosine triphosphate, the tri-phosphorylated metabolite of GCV (GCV-TP) is preferentially incorporated by the viral DNA polymerase, thereby terminating chain extension and, eventually, viral replication. However, the treatment outcome of GCV varies greatly among individuals, therefore warranting better understanding of its metabolism. Here we show that NUDT15, a Nudix hydrolase known to metabolize thiopurine triphosphates, can similarly hydrolyze GCV-TP through biochemical studies and co-crystallization of the NUDT15/GCV-TP complex. More critically, GCV efficacy was potentiated in HCMV-infected cells following NUDT15 depletion by RNAi or inhibition by an in-house-developed, nanomolar NUDT15 inhibitor, TH8321, suggesting that pharmacological targeting of NUDT15 is a possible avenue to improve existing anti-HCMV regimens. Collectively, the data further implicate NUDT15 as a broad-spectrum metabolic regulator of nucleoside analog therapeutics, such as thiopurines and GCV.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Ganciclovir/pharmacology , Pyrophosphatases/metabolism , Antiviral Agents/chemistry , Cell Line, Tumor , Female , Ganciclovir/chemistry , Humans , Hydrolysis , Microbial Sensitivity Tests , Recombinant Proteins/metabolism
3.
Oncol Rep ; 41(5): 2927-2936, 2019 May.
Article in English | MEDLINE | ID: mdl-30896862

ABSTRACT

Glioblastoma (GBM) is the most aggressive form of brain tumor in adults, with a devastating outcome. Emerging evidence shows that human cytomegalovirus (HCMV) proteins and nucleic acids are present in GBM tissues. DNA methylation is important for the initiation and progression of cancer and is an established host response against invading nucleic acids. The expression and localization of DNA methyltransferase 1 (DNMT­1) was assessed, and the effects of DNA methylation inhibitor 5­azacytidine (5AZA) were analyzed in the context of the viral replication, proliferation and invasion capacities of HCMV­infected GBM U343MG cells. In addition, the expression of various HCMV proteins and DNMT­1 was examined in GBM tissue specimens obtained from five patients. DNMT­1 was localized in the nucleus of cells expressing HCMV­immediate early, whereas in cells expressing HCMV­glycoprotein gB (gB), extranuclear/cytoplasmic localization was observed. This was also observed in vitro in U343MG cells. In addition, DNMT­1 was localized to the extranuclear/cytoplasmic space of cells lining blood vessel walls within the GBM tumors. Treatment of infected U343MG cells with 5AZA did not affect viral replication, but reduced cell invasion and proliferation (P=0.05 and P<0.0001, respectively). However, 5AZA treatment of uninfected cells did not affect cell invasion (P=0.09), but proliferation was significantly reduced (P<0.0001). These findings may be of importance in further investigations aimed at using DNA methylation and viral inhibitors in GBM therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Brain Neoplasms/drug therapy , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Glioblastoma/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/virology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cytomegalovirus/drug effects , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Cytoplasm/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA Methylation/drug effects , Disease Progression , Female , Glioblastoma/pathology , Glioblastoma/virology , Humans , Male , Middle Aged , Treatment Outcome , Viral Envelope Proteins/metabolism , Virus Replication/drug effects
4.
Int J Mol Sci ; 20(3)2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30764543

ABSTRACT

Colorectal cancer is the source of one of the most common cancer-related deaths worldwide, where the main cause of patient mortality remains metastasis. The aim of this study was to determine the role of CCL7 (chemokine (C-C motif) ligand 7) in tumor progression and finding whether it could predict survival of colorectal cancer patients. Initially, our study focused on the crosstalk between mesenchymal stem cells (MSCs) and CT26 colon carcinoma cells and resulted in identifying CCL7 as a chemokine upregulated in CT26 colon cancer cells cocultured with MSCs, compared with CT26 in monoculture in vitro. Moreover, we showed that MSCs enhance CT26 tumor cell proliferation and migration. We analyzed the effect of CCL7 overexpression on tumor progression in a murine CT26 model, where cells overexpressing CCL7 accelerated the early phase of tumor growth and caused higher lung metastasis rates compared with control mice. Microarray analysis revealed that tumors overexpressing CCL7 had lower expression of immunoglobulins produced by B lymphocytes. Additionally, using Jh mutant mice, we confirmed that in the CT26 model, CCL7 has an immunoglobulin-, and thereby, B-cell-dependent effect on metastasis formation. Finally, higher expression of CCL7 receptor CCR2 (C-C chemokine receptor type 2) was associated with shorter overall survival of colorectal cancer patients. Altogether, we showed that CCL7 is essentially involved in the progression of colorectal cancer in a CT26 mouse model and that the expression of its receptor CCR2 could be related to a different outcome pattern of patients with colorectal carcinoma.


Subject(s)
Chemokine CCL7/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Receptors, CCR2/genetics , Up-Regulation , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CCL7/analysis , Colonic Neoplasms/pathology , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Receptors, CCR2/analysis
5.
BMC Cancer ; 18(1): 154, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29409474

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common malignant brain tumor with median survival of 12-15 months. Owing to uncertainty in clinical outcome, additional prognostic marker(s) apart from existing markers are needed. Since overexpression of endothelin B receptor (ETBR) has been demonstrated in gliomas, we aimed to test whether ETBR is a useful prognostic marker in GBM and examine if the clinically available endothelin receptor antagonists (ERA) could be useful in the disease treatment. METHODS: Data from The Cancer Genome Atlas and the Gene Expression Omnibus database were analyzed to assess ETBR expression. For survival analysis, glioblastoma samples from 25 Swedish patients were immunostained for ETBR, and the findings were correlated with clinical history. The druggability of ETBR was assessed by protein-protein interaction network analysis. ERAs were analyzed for toxicity in in vitro assays with GBM and breast cancer cells. RESULTS: By bioinformatics analysis, ETBR was found to be upregulated in glioblastoma patients, and its expression levels were correlated with reduced survival. ETBR interacts with key proteins involved in cancer pathogenesis, suggesting it as a druggable target. In vitro viability assays showed that ERAs may hold promise to treat glioblastoma and breast cancer. CONCLUSIONS: ETBR is overexpressed in glioblastoma and other cancers and may be a prognostic marker in glioblastoma. ERAs may be useful for treating cancer patients.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Receptor, Endothelin B/genetics , Adult , Aged , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Endothelin Receptor Antagonists/therapeutic use , Female , Gene Regulatory Networks , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Molecular Targeted Therapy , Prognosis , Receptor, Endothelin B/metabolism
6.
Int J Cancer ; 142(6): 1266-1276, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29067692

ABSTRACT

Glioma stem cells (GSCs) are glioblastoma (GBM) cells that are resistant to therapy and can give rise to recurrent tumors. The identification of patient-related factors that support GSCs is thus necessary to design effective therapies for GBM patients. Glucocorticoids (GCs) are used to treat GBM-associated edema. However, glucocorticoids participate in the physiological response to psychosocial stress, which has been linked to poor cancer prognosis. This raises concern that glucocorticoids affect the tumor and GSCs. Here, we treated primary human GBM cells with dexamethasone and evaluated GC-driven changes in cell morphology, proliferation, migration, gene expression, secretory activity and growth as neurospheres. Dexamethasone treatment of GBM cells appeared to promote the development of a GSC-like phenotype and conferred resistance to physiological stress and chemotherapy. We also analyzed a potential correlation between GC treatment and tumor recurrence after surgical excision in a population-based consecutive cohort of 48 GBM patients, adjusted for differences in known prognostic factors concerning baseline and treatment characteristics. In this cohort, we found a negative correlation between GC intake and progression-free survival, regardless of the MGMT methylation status. In conclusion, our findings raise concern that treatment of GBM with GCs may compromise the efficacy of chemotherapy and may support a GSC population, which could contribute to tumor recurrence and the poor prognosis of the disease.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Drug Resistance, Neoplasm/drug effects , Glioblastoma/therapy , Glucocorticoids/adverse effects , Neoplasm Recurrence, Local/prevention & control , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Brain/pathology , Brain/surgery , Brain Edema/drug therapy , Brain Edema/etiology , Brain Neoplasms/complications , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Chemoradiotherapy, Adjuvant/methods , Dexamethasone/adverse effects , Disease-Free Survival , Female , Glioblastoma/complications , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neurosurgical Procedures , Primary Cell Culture , Prognosis , Stress, Physiological/drug effects , Treatment Outcome , Tumor Cells, Cultured
7.
J Gen Virol ; 98(5): 1058-1072, 2017 May.
Article in English | MEDLINE | ID: mdl-28589873

ABSTRACT

Human cytomegalovirus (HCMV) infection results in the production of virions, dense bodies (DBs) and non-infectious enveloped particles, all of which incorporate proteins and RNAs that can be transferred to host cells. Here, we investigated whether virions and DBs also carry microRNAs (miRNAs) and assessed their delivery and functionality in cells. Human lung fibroblasts (MRC-5) were infected with the HCMV strain AD169, and conditioned cell culture medium was collected and centrifuged. The pellets were treated with RNase-ONE, and the virions and DBs were purified with a potassium tartrate-glycerol gradient and dialysed. The virions and DBs were incubated with micrococcal nuclease, DNA and RNA were extracted and then analysed with TaqMan PCR assays, while the proteins were examined with Western blots. To assess the delivery of miRNAs to cells and their functionality, virions and DBs were irradiated with UV light. The purity of the virions and DBs was confirmed by typical morphology, the presence of the structural protein pp65 and the HCMV genome, the ability to infect MRC-5 cells and the absence of the host genome. RNA analysis revealed the presence of 14 HCMV-encoded miRNAs (UL22A-5p, US25-1-5p, UL22A-3p, US5-2-3p, UL112-3p, US25-2-3p, US25-2-5p, US33-3p, US5-1, UL36-5p, US4-5p, UL36-3p, UL70-5p and US25-1-3p), HCMV immediate-early mRNA and long non-coding RNA2.7, moreover, two host-encoded miRNAs (hsa-miR-218-5p and hsa-miR-21-5p) and beta-2-microglobulin RNA. UV-irradiated virions and DBs delivered viral miRNAs (US25-1-5p and UL112-3p) to the host cells, and miR-US25-1-5p was functional in a luciferase reporter assay. We conclude that virions and DBs carry miRNAs that are biologically functional and can be delivered to cells, which may affect cellular processes.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/metabolism , MicroRNAs/metabolism , RNA, Viral/metabolism , Virion/metabolism , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Host-Pathogen Interactions , Humans , MicroRNAs/genetics , RNA, Viral/genetics , Virion/genetics
8.
Elife ; 62017 06 22.
Article in English | MEDLINE | ID: mdl-28639941

ABSTRACT

Staff from the Mayo Clinic in the US and the Karolinska Institute in Sweden describe a joint transatlantic course intended to broaden the horizons of the next generation of researchers in the field of regenerative medicine.


Subject(s)
Biomedical Research/methods , Education/methods , International Cooperation , Students , Creativity , Social Behavior , Sweden
9.
Oncotarget ; 7(48): 79572-79583, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27788487

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in humans and is characterized with poor outcome. In this study, we investigated components of prolactin (Prl) system in cell models of GBM and in histological tissue sections obtained from GBM patients. Expression of Prolactin receptor (PrlR) was detected at high levels in U251-MG, at low levels in U87-MG and barely detectable in U373 cell lines and in 66% of brain tumor tissues from 32 GBM patients by immunohistochemical technique. In addition, stimulation of U251-MG and U87-MG cells but not U373 with Prl resulted in increased STAT5 phosphorylation and only in U251-MG cells with increased cellular invasion. Furthermore, STAT5 phosphorylation and cellular invasion induced in Prl stimulated cells were significantly reduced by using a Prl receptor antagonist that consists of Prl with four amino acid replacements. We conclude that Prl receptor is expressed at different levels in the majority of GBM tumors and that blocking of PrlR in U251-MG cells significantly reduce cellular invasion.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement/drug effects , Glioblastoma/metabolism , Prolactin/pharmacology , Receptors, Prolactin/agonists , STAT5 Transcription Factor/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Glioblastoma/drug therapy , Glioblastoma/pathology , Hormone Antagonists/pharmacology , Humans , Neoplasm Invasiveness , Phosphorylation , Receptors, Prolactin/antagonists & inhibitors , Receptors, Prolactin/metabolism , Signal Transduction/drug effects
10.
J Tissue Eng Regen Med ; 10(3): 222-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-23281223

ABSTRACT

Stem cell-based therapies hold great promise as a clinically viable approach for vascular regeneration. Preclinical studies have been very encouraging and early clinical trials have suggested favourable outcomes. However, significant challenges remain in terms of optimizing cell retention and maintenance of the paracrine effects of implanted cells. To address these issues, we have proposed the use of a cellular encapsulation approach to enhance vascular regeneration. We contained human mesenchymal stem cells (hMSCs) in biocompatible alginate microcapsules for therapeutic treatment in the setting of murine hindlimb ischaemia. This approach supported the paracrine pro-angiogenic activity of hMSCs, prevented incorporation of hMSCs into the host tissue and markedly enhanced their therapeutic effect. While injection of non-encapsulated hMSCs resulted in a 22 ± 10% increase in vascular density and no increase in perfusion, treatment with encapsulated hMSCs resulted in a 70 ± 8% increase in vascular density and 21 ± 7% increase in perfusion. The described cellular encapsulation strategy may help to better define the mechanisms responsible for the beneficial effects of cell-based therapies and provide a therapeutic strategy for inducing vascular growth in the adult. As hMSCs are relatively easy to isolate from patients, and alginate is biocompatible and already used in clinical applications, therapeutic cell encapsulation for vascular repair represents a highly translatable platform for cell-based therapy in humans.


Subject(s)
Alginates/pharmacology , Hindlimb/blood supply , Ischemia/therapy , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Paracrine Communication/drug effects , Animals , Capsules , Cell Survival/drug effects , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Hindlimb/drug effects , Hindlimb/pathology , Humans , Ischemia/pathology , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mice, Nude , Permeability , Wound Healing/drug effects
11.
Arterioscler Thromb Vasc Biol ; 34(7): 1548-55, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24855063

ABSTRACT

OBJECTIVE: Collateral vessel formation can functionally compensate for obstructive vascular lesions in patients with atherosclerosis. Neovascularization processes are triggered by fluid shear stress, hypoxia, growth factors, chemokines, proteases, and inflammation, as well as reactive oxygen species, in response to ischemia. Polymerase δ-interacting protein 2 (Poldip2) is a multifunctional protein that regulates focal adhesion turnover and vascular smooth muscle cell migration and modifies extracellular matrix composition. We, therefore, tested the hypothesis that loss of Poldip2 impairs collateral formation. APPROACH AND RESULTS: The mouse hindlimb ischemia model has been used to understand mechanisms involved in postnatal blood vessel formation. Poldip2(+/-) mice were subjected to femoral artery excision, and functional and morphological analysis of blood vessel formation was performed after injury. Heterozygous deletion of Poldip2 decreased the blood flow recovery and spontaneous running activity at 21 days after injury. H2O2 production, as well as the activity of matrix metalloproteinases-2 and -9, was reduced in these animals compared with Poldip2(+/+) mice. Infiltration of macrophages in the peri-injury muscle was also decreased; however, macrophage phenotype was similar between genotypes. In addition, the formation of capillaries and arterioles was impaired, as was angiogenesis, in agreement with a decrease in proliferation observed in endothelial cells treated with small interfering RNA against Poldip2. Finally, regression of newly formed vessels and apoptosis was more pronounced in Poldip2(+/-) mice. CONCLUSIONS: Together, these results suggest that Poldip2 promotes ischemia-induced collateral vessel formation via multiple mechanisms that likely involve reactive oxygen species-dependent activation of matrix metalloproteinase activity, as well as enhanced vascular cell growth and survival.


Subject(s)
Ischemia/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Nuclear Proteins/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Collateral Circulation , Disease Models, Animal , Heterozygote , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hydrogen Peroxide/metabolism , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Macrophages/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , RNA Interference , Recovery of Function , Regional Blood Flow , Time Factors , Transfection
12.
J Am Heart Assoc ; 2(5): e000367, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24113327

ABSTRACT

BACKGROUND: Stem cells for cardiac repair have shown promise in preclinical trials, but lower than expected retention, viability, and efficacy. Encapsulation is one potential strategy to increase viable cell retention while facilitating paracrine effects. METHODS AND RESULTS: Human mesenchymal stem cells (hMSC) were encapsulated in alginate and attached to the heart with a hydrogel patch in a rat myocardial infarction (MI) model. Cells were tracked using bioluminescence (BLI) and cardiac function measured by transthoracic echocardiography (TTE) and cardiac magnetic resonance imaging (CMR). Microvasculature was quantified using von Willebrand factor staining and scar measured by Masson's Trichrome. Post-MI ejection fraction by CMR was greatly improved in encapsulated hMSC-treated animals (MI: 34 ± 3%, MI + Gel: 35 ± 3%, MI + Gel + hMSC: 39 ± 2%, MI + Gel + encapsulated hMSC: 56 ± 1%; n = 4 per group; P < 0.01). Data represent mean ± SEM. By TTE, encapsulated hMSC-treated animals had improved fractional shortening. Longitudinal BLI showed greatest hMSC retention when the cells were encapsulated (P < 0.05). Scar size at 28 days was significantly reduced in encapsulated hMSC-treated animals (MI: 12 ± 1%, n = 8; MI + Gel: 14 ± 2%, n = 7; MI + Gel + hMSC: 14 ± 1%, n = 7; MI+Gel+encapsulated hMSC: 7 ± 1%, n = 6; P < 0.05). There was a large increase in microvascular density in the peri-infarct area (MI: 121 ± 10, n = 7; MI + Gel: 153 ± 26, n = 5; MI + Gel + hMSC: 198 ± 18, n = 7; MI + Gel + encapsulated hMSC: 828 ± 56 vessels/mm2, n = 6; P < 0.01). CONCLUSIONS: Alginate encapsulation improved retention of hMSCs and facilitated paracrine effects such as increased peri-infarct microvasculature and decreased scar. Encapsulation of MSCs improved cardiac function post-MI and represents a new, translatable strategy for optimization of regenerative therapies for cardiovascular diseases.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/surgery , Animals , Cell Culture Techniques/methods , Graft Survival , Humans , Male , Rats
13.
Small ; 9(23): 4017-26, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-23766267

ABSTRACT

Cell therapies offer exciting new opportunities for effectively treating many human diseases. However, delivery of therapeutic cells by intravenous injection, while convenient, relies on the relatively inefficient process of homing of cells to sites of injury. To address this limitation, a novel strategy has been developed to load cells with superparamagnetic iron oxide nanoparticles (SPIOs), and to attract them to specific sites within the body by applying an external magnetic field. The feasibility of this approach is demonstrated using human mesenchymal stem cells (hMSCs), which may have a significant potential for regenerative cell therapies due to their ease of isolation from autologous tissues, and their ability to differentiate into various lineages and modulate their paracrine activity in response to the microenvironment. The efficient loading of hMSCs with polyethylene glycol-coated SPIOs is achieved, and it is found that SPIOs are localized primarily in secondary lysosomes of hMSCs and are not toxic to the cells. Further, the key stem cell characteristics, including the immunophenotype of hMSCs and their ability to differentiate, are not altered by SPIO loading. Through both experimentation and mathematical modeling, it is shown that, under applied magnetic field gradients, SPIO-containing cells can be localized both in vitro and in vivo. The results suggest that, by loading SPIOs into hMSCs and applying appropriate magnetic field gradients, it is possible to target hMSCs to particular vascular networks.


Subject(s)
Ferric Compounds/chemistry , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cells/cytology , Nanoparticles/chemistry , Humans , Magnetite Nanoparticles/adverse effects , Nanoparticles/adverse effects
14.
Atherosclerosis ; 228(1): 168-74, 2013 May.
Article in English | MEDLINE | ID: mdl-23466073

ABSTRACT

OBJECTIVE: Osteopontin (OPN) is a multifunctional protein found in abundance in atherosclerotic plaques. Angiotensin II (Ang II) promotes atherosclerosis by inducing adhesion and migration of vascular smooth muscle cells (VSMCs). MicroRNAs (miRNAs) are critical regulators of protein expression. However, the relationship between Ang II, miRNAs and OPN has yet to be fully explored. METHODS AND RESULTS: Using cultured VSMCs, we found that Ang II increased cellular OPN protein expression 4 h after treatment by 420 ± 54% (p < 0.03) in a translation dependent manner. Sequence analysis revealed a putative binding site for mir181a and raised the possibility that miR181a is a potential regulatory mechanism for OPN expression. We demonstrated that Ang II decreased miR181a expression by 52 ± 7% (p < 0 .0001) and overexpressing miR181a inhibited Ang II induced increases in OPN protein expression by 69 ± 9% (p < 0.05). Furthermore, we demonstrated that miR181a is functionally important in that overexpression of miR181a inhibited VSMCs adhesion to collagen in response to Ang II as compared to controls by 36 ± 4%. (p < 0.05) CONCLUSIONS: These results demonstrate that miR181a regulates OPN expression and that altering miR181a expression may be a novel therapeutic approach to modulate OPN protein expression.


Subject(s)
Angiotensin II/pharmacology , MicroRNAs/genetics , Muscle, Smooth, Vascular/physiology , Osteopontin/genetics , Animals , Aorta/cytology , Cells, Cultured , Gene Expression/drug effects , Gene Expression/physiology , MicroRNAs/pharmacology , Muscle, Smooth, Vascular/cytology , RNA, Messenger/metabolism , Rats , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Vasoconstrictor Agents/pharmacology
15.
Am J Physiol Regul Integr Comp Physiol ; 303(1): R48-56, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22573106

ABSTRACT

The formation of vascular networks during embryogenesis and early stages of development encompasses complex and tightly regulated growth of blood vessels, followed by maturation of some vessels, and spatially controlled disconnection and pruning of others. The adult vasculature, while more quiescent, is also capable of adapting to changing physiological conditions by remodeling blood vessels. Numerous studies have focused on understanding key factors that drive vessel growth in the adult in response to ischemic injury. However, little is known about the extent of vessel rarefaction and its potential contribution to the final outcome of vascular recovery. We addressed this topic by characterizing the endogenous phases of vascular repair in a mouse model of hindlimb ischemia. We showed that this process is biphasic. It encompasses an initial rapid phase of vessel growth, followed by a later phase of vessel rarefaction. In healthy mice, this process resulted in partial recovery of perfusion and completely restored the ability of mice to run voluntarily. Given that the ability to revascularize can be compromised by a cardiovascular risk factor such as diabetes, we also examined vascular repair in diabetic mice. We found that paradoxically both the initial growth and subsequent regression of collateral vessels were more pronounced in the setting of diabetes and resulted in impaired recovery of perfusion and impaired functional status. In conclusion, our findings demonstrate that the formation of functional collateral vessels in the hindlimb requires vessel growth and subsequent vessel rarefaction. In the setting of diabetes, the physiological defect was not in the initial formation of vessels but rather in the inability to sustain newly formed vessels.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Hindlimb/blood supply , Ischemia/physiopathology , Neovascularization, Physiologic/physiology , Animals , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Laser-Doppler Flowmetry , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Regional Blood Flow/physiology , Streptozocin/adverse effects
16.
Arterioscler Thromb Vasc Biol ; 32(6): 1383-91, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492090

ABSTRACT

OBJECTIVE: Previous findings from our laboratory demonstrated that neovascularization was impaired in osteopontin (OPN) knockout animals. However, the mechanisms responsible for the regulation of OPN expression in the setting of ischemia remain undefined. Therefore, we sought to determine whether OPN is upregulated in response to ischemia and hypothesized that hydrogen peroxide (H(2)O(2)) is a critical component of the signaling mechanism by which OPN expression is upregulated in response to ischemia in vivo. METHODS AND RESULTS: To determine whether ischemic injury upregulates OPN, we used a murine model of hindlimb ischemia. Femoral artery ligation in C57BL/6 mice significantly increased OPN expression and H(2)O(2) production. Infusion of C57BL/6 mice with polyethylene glycol-catalase (10 000 U/kg per day) or the use of transgenic mice with smooth muscle cell-specific catalase overexpression blunted ischemia-induced OPN, suggesting ischemia-induced OPN expression is H(2)O(2)-dependent. Decreased H(2)O(2)-mediated OPN blunted reperfusion and collateral formation in vivo. In contrast, the overexpression of OPN using lentivirus restored neovascularization. CONCLUSIONS: Scavenging H(2)O(2) blocks ischemia-induced OPN expression, providing evidence that ischemia-induced OPN expression is H(2)O(2) dependent. Decreased OPN expression impaired neovascularization, whereas overexpression of OPN increased angiogenesis, supporting our hypothesis that OPN is a critical mediator of postischemic neovascularization and a potential novel therapeutic target for inducing new vessel growth.


Subject(s)
Hydrogen Peroxide/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Osteopontin/metabolism , Oxidative Stress , Animals , Antioxidants/administration & dosage , Catalase/administration & dosage , Catalase/genetics , Catalase/metabolism , Cells, Cultured , Collateral Circulation , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors , Hindlimb , Humans , Infusions, Intravenous , Ischemia/diagnostic imaging , Ischemia/genetics , Ischemia/physiopathology , Laser-Doppler Flowmetry , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Neovascularization, Physiologic/drug effects , Osteopontin/genetics , Oxidative Stress/drug effects , Polyethylene Glycols/administration & dosage , Regional Blood Flow , Signal Transduction , Time Factors , Up-Regulation , X-Ray Microtomography
18.
Arterioscler Thromb Vasc Biol ; 31(10): 2203-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21799178

ABSTRACT

OBJECTIVE: Myeloid lineage cells (MLCs) such as macrophages are known to play a key role in postischemic neovascularization. However, the role of MLC-derived reactive oxygen species in this process and their specific chemical identity remain unknown. METHODS AND RESULTS: Transgenic mice with MLC-specific overexpression of catalase (Tg(Cat-MLC) mice) were created on a C57BL/6 background. Macrophage catalase activity was increased 3.4-fold compared with wild-type mice. After femoral artery ligation, laser Doppler perfusion imaging revealed impaired perfusion recovery in Tg(Cat-MLC) mice. This was associated with fewer collateral vessels, as assessed by microcomputed tomography angiography, and decreased capillary density. Impaired functional recovery of the ischemic limb was also evidenced by a 50% reduction in spontaneous running activity. The deficient neovascularization was associated with a blunted inflammatory response, characterized by decreased macrophage infiltration of ischemic tissues, and lower mRNA levels of inflammatory markers, such as tumor necrosis factor-α, osteopontin, and matrix mettaloproteinase-9. In vitro macrophage migration was impaired in Tg(Cat-MLC) mice, suggesting a role for H(2)O(2) in regulating the ability of macrophages to infiltrate ischemic tissues. CONCLUSIONS: MLC-derived H(2)O(2) plays a key role in promoting neovascularization in response to ischemia and is a necessary factor for the development of ischemia-induced inflammation.


Subject(s)
Capillaries/enzymology , Catalase/biosynthesis , Hydrogen Peroxide/metabolism , Ischemia/enzymology , Muscle, Skeletal/blood supply , Myeloid Cells/enzymology , Neovascularization, Physiologic , Animals , Capillaries/diagnostic imaging , Capillaries/physiopathology , Catalase/genetics , Cell Movement , Cells, Cultured , Collateral Circulation , Disease Models, Animal , Endothelial Cells/metabolism , Femoral Artery/surgery , Genotype , Hindlimb , Humans , Inflammation Mediators/metabolism , Ischemia/genetics , Ischemia/physiopathology , Laser-Doppler Flowmetry , Ligation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Neovascularization, Physiologic/genetics , Phenotype , RNA, Messenger/metabolism , Regional Blood Flow , Stem Cells/metabolism , Time Factors , Ultrasonography , Up-Regulation , X-Ray Microtomography
19.
Proc Natl Acad Sci U S A ; 107(8): 3323-8, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20080569

ABSTRACT

Therapeutic vascularization remains a significant challenge in regenerative medicine applications. Whether the goal is to induce vascular growth in ischemic tissue or scale up tissue-engineered constructs, the ability to induce the growth of patent, stable vasculature is a critical obstacle. We engineered polyethylene glycol-based bioartificial hydrogel matrices presenting protease-degradable sites, cell-adhesion motifs, and growth factors to induce the growth of vasculature in vivo. Compared to injection of soluble VEGF, these matrices delivered sustained in vivo levels of VEGF over 2 weeks as the matrix degraded. When implanted subcutaneously in rats, degradable constructs containing VEGF and arginine-glycine-aspartic acid tripeptide induced a significant number of vessels to grow into the implant at 2 weeks with increasing vessel density at 4 weeks. The mechanism of enhanced vascularization is likely cell-demanded release of VEGF, as the hydrogels may degrade substantially within 2 weeks. In a mouse model of hind-limb ischemia, delivery of these matrices resulted in significantly increased rate of reperfusion. These results support the application of engineered bioartificial matrices to promote vascularization for directed regenerative therapies.


Subject(s)
Neovascularization, Physiologic/drug effects , Pharmaceutical Vehicles/chemistry , Regeneration/drug effects , Regenerative Medicine/methods , Animals , Disease Models, Animal , Extremities/blood supply , Hydrogels/administration & dosage , Hydrogels/chemistry , Ischemia/therapy , Male , Mice , Mice, Inbred Strains , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Pharmaceutical Vehicles/administration & dosage , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Rats , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL