Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Circ J ; 84(7): 1183-1188, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32522903

ABSTRACT

BACKGROUND: NT5Egenetic mutations are known to result in calcification of joints and arteries (CALJA), and worldwide, 14 patients from 7 families have been reported.Methods and Results:A total of 5 patients from 2 independent families with CALJA were found in Japan. Of them, 3 complained of intermittent claudication (IC), and 1 suffered from bilateral chronic limb-threatening ischemia (CLTI). Whole-exome sequencing analysis revealed an identical mutation pattern (c.G3C on the exon 1 start codon) that was unique compared withNT5Emutations reported in other countries. CONCLUSIONS: Vascular specialists need to recognize CALJA as a rare cause of ischemic IC and CLTI.


Subject(s)
5'-Nucleotidase/genetics , Calcinosis/genetics , Intermittent Claudication/genetics , Ischemia/genetics , Joint Diseases/genetics , Mutation , Vascular Calcification/genetics , Vascular Diseases/genetics , Adult , Aged , Aged, 80 and over , Calcinosis/diagnosis , Calcinosis/enzymology , Chronic Disease , Exons , Female , GPI-Linked Proteins/genetics , Genetic Predisposition to Disease , Humans , Intermittent Claudication/diagnosis , Intermittent Claudication/enzymology , Ischemia/diagnosis , Ischemia/enzymology , Joint Diseases/diagnosis , Joint Diseases/enzymology , Male , Middle Aged , Phenotype , Vascular Calcification/diagnostic imaging , Vascular Calcification/enzymology , Vascular Diseases/diagnosis , Vascular Diseases/enzymology , Exome Sequencing
2.
Vasc Med ; 25(5): 401-410, 2020 10.
Article in English | MEDLINE | ID: mdl-32853041

ABSTRACT

Flow-limiting atherosclerotic lesions of arteries supplying the limbs are a cause of symptoms in patients with peripheral artery disease (PAD). Musculoskeletal metabolic factors also contribute to the pathophysiology of claudication, which is manifest as leg discomfort that impairs walking capacity. Accordingly, we conducted a case-control study to determine whether skeletal muscle metabolic gene expression is altered in PAD. Calf skeletal muscle gene expression of patients with PAD and healthy subjects was analyzed using microarrays. The top-ranking gene differentially expressed between PAD and controls (FDR < 0.001) was PLA2G16, which encodes adipose-specific phospholipase A2 (AdPLA) and is implicated in the maintenance of insulin sensitivity and regulation of lipid metabolism. Differential expression was confirmed by qRT-PCR; PLA2G16 was downregulated by 68% in patients with PAD (p < 0.001). Expression of Pla2g16 was then measured in control (db/+) and diabetic (db/db) mice that underwent unilateral femoral artery ligation. There was significantly reduced expression of Pla2g16 in the ischemic leg of both control and diabetic mice (by 51%), with significantly greater magnitude of reduction in the diabetic mice (by 79%). We conclude that AdPLA is downregulated in humans with PAD and in mice with hindlimb ischemia. Reduced AdPLA may contribute to impaired walking capacity in patients with PAD via its effects on skeletal muscle metabolism. Further studies are needed to fully characterize the role of AdPLA in PAD and to investigate its potential as a therapeutic target for alleviating symptoms of claudication.


Subject(s)
Intermittent Claudication/enzymology , Ischemia/enzymology , Muscle, Skeletal/enzymology , Peripheral Arterial Disease/enzymology , Phospholipases A2, Calcium-Independent/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Animals , Case-Control Studies , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , Humans , Insulin Resistance , Intermittent Claudication/genetics , Intermittent Claudication/physiopathology , Ischemia/genetics , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Middle Aged , Muscle, Skeletal/physiopathology , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/physiopathology , Phospholipases A2, Calcium-Independent/genetics , Tumor Suppressor Proteins/genetics , Walking
3.
Vasc Med ; 21(1): 21-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26584888

ABSTRACT

UNLABELLED: Alternative treatment strategies for claudication are needed and cell-based therapies designed to induce angiogenesis are promising. The purpose of this report was to conduct a Phase I safety, dose-escalating, non-randomized, open-label study of autologous, fully differentiated venous endothelial and smooth muscle cells called MultiGeneAngio (MGA) for claudication due to peripheral artery disease. Twelve subjects, at two centers, received a single intra-arterial infusion of a suspension of equal amounts of transduced autologous venous smooth muscle cells expressing vascular endothelial growth factor (VEGF165) and endothelial cells expressing angiopoietin-1 (Ang-1) (Cohort 1: 1 × 10(7), Cohort 2: 2 × 10(7), Cohort 3: 5 × 10(7), Cohort 4: 7 × 10(7)). The treatment was given unblinded and in the more symptomatic lower extremity. Transduced cells were tested for in vitro doubling time, telomerase activity, and gene expression. The main outcomes were clinical safety and tolerability. Other safety measures included ankle-brachial index (ABI) and walking time on a treadmill. All subjects were male (mean age 60 ± 5 years) including 25% with diabetes mellitus. At 1-year follow-up, there was one serious adverse event possibly related to MGA. Safety endpoints including VEGF and Ang-1 plasma protein levels were within normal ranges in all subjects. The mean maximal walking time increased from baseline to 1 year and the index limb ABI was unchanged, indicating no safety concerns. MGA, an autologous, transduced, cell-based therapy was well tolerated and safe in this Phase I study. Further evaluation is warranted in randomized human studies. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT00390767.


Subject(s)
Angiogenic Proteins/biosynthesis , Cell Transplantation/methods , Endothelial Cells/transplantation , Genetic Therapy/methods , Intermittent Claudication/surgery , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Peripheral Arterial Disease/surgery , Aged , Angiogenic Proteins/genetics , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Ankle Brachial Index , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Exercise Test , Exercise Tolerance , Humans , Intermittent Claudication/diagnosis , Intermittent Claudication/genetics , Intermittent Claudication/metabolism , Intermittent Claudication/physiopathology , Male , Michigan , Middle Aged , Myocytes, Smooth Muscle/metabolism , Pennsylvania , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology , Recovery of Function , Telomerase/metabolism , Time Factors , Transduction, Genetic , Treatment Outcome , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
4.
J Vasc Surg ; 61(1): 155-61, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25095746

ABSTRACT

OBJECTIVE: Inflammation contributes to the development of peripheral artery disease (PAD) and may contribute to intermittent claudication by adversely affecting vascular and skeletal muscle function. We explored the association of inflammation to maximal walking time (MWT) in patients with claudication. METHODS: Circulating inflammatory biomarkers, including tumor necrosis factor α (TNF-α), C-reactive protein (CRP), interleukin-6 (IL-6), and soluble intercellular adhesion molecule 1 (sICAM), were measured in 75 subjects with intermittent claudication as well as in 43 healthy subjects. Real-time polymerase chain reaction was used to quantify mRNA expression of TNF-α, IL-6, interferon-γ, and CD36 from peripheral blood monocytes. Treadmill testing was performed in PAD subjects to assess MWT. RESULTS: Compared with healthy subjects, PAD subjects had higher levels of circulating TNF-α (P < .0001), CRP (P = .003), sICAM (P < .0001), and IL-6 (P < .0001). Expression of both IL-6 (P = .024) and CD36 (P = .018) was greater in PAD subjects than in healthy subjects. Among subjects with PAD, higher gene expression of TNF-α was associated inversely with MWT (P = .01). MWT was also associated inversely with greater levels of circulating TNF-α (P = .028), CRP (P = .024), IL-6 (P = .03), and sICAM (P = .018). CONCLUSIONS: Systemic inflammation, as indicated by TNF-α inflammatory gene expression in peripheral blood monocytes and by circulating biomarker levels, is associated with impairment in walking time in patients with PAD and intermittent claudication.


Subject(s)
Inflammation Mediators/blood , Intermittent Claudication/diagnosis , Monocytes/metabolism , Peripheral Arterial Disease/diagnosis , Tumor Necrosis Factor-alpha/blood , Walking , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Exercise Test , Exercise Tolerance , Female , Humans , Intermittent Claudication/blood , Intermittent Claudication/genetics , Intermittent Claudication/immunology , Intermittent Claudication/physiopathology , Male , Middle Aged , Peripheral Arterial Disease/blood , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/immunology , Peripheral Arterial Disease/physiopathology , Predictive Value of Tests , RNA, Messenger/blood , Real-Time Polymerase Chain Reaction , Severity of Illness Index , Time Factors , Tumor Necrosis Factor-alpha/genetics
5.
Eur J Vasc Endovasc Surg ; 50(4): 494-501, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26122834

ABSTRACT

OBJECTIVE: Critical limb ischemia (CLI) is the most severe form of peripheral arterial disease and a major unmet public health care need. This phase I clinical study was performed to assess the safety and preliminary efficacy of naked plasmid DNA (pUDK-HGF) expressing human hepatocyte growth factor (HGF) in patients with critical limb ischemia (CLI). DESIGN: Twenty-one patients with CLI were enrolled and randomly divided into four dose groups (4-16 mg) to receive local injection of pUDK-HGF into ischemic calf and/or thigh muscles twice on days 1 and 15. Safety, including adverse events and physiological parameters, and preliminary efficacy, including pain severity score (VAS), ulcer size, transcutaneous oxygen pressure (TcPO2), and ankle brachial index (ABI), were evaluated throughout a 3 month follow up period. RESULTS: All doses of pUDK-HGF were well tolerated by the patients. None of the adverse effects was considered to be related to pUDK-HGF injection. Two significant clinical results were observed after pUDK-HGF administration. The mean VAS value of all patients decreased from 4.52 at baseline to 0.30 (p < .01), and pain had disappeared in 14 out of 17 evaluable patients by day 91. Two of four ulcers had completely healed, with the other two patients having more than 25% ulcer size reduction in the long axis diameter. Of five patients with gangrene, one gangrenous wound had healed completely and two patients showed marked size reduction by day 91. The mean hemodynamic parameters (ABI, TcPO2) were also improved. CONCLUSION: Intramuscular injection of pUDK-HGF is safe, and may provide symptomatic relief for CLI patients. A larger, randomized, double blinded phase II trial will provide more information on safety and efficacy.


Subject(s)
Genetic Therapy/methods , Hepatocyte Growth Factor/biosynthesis , Ischemia/therapy , Lower Extremity/blood supply , Peripheral Arterial Disease/therapy , Adult , Aged , Ankle Brachial Index , Blood Gas Monitoring, Transcutaneous , China , Critical Illness , Female , Gene Transfer Techniques , Genetic Therapy/adverse effects , Hemodynamics , Hepatocyte Growth Factor/genetics , Humans , Injections, Intramuscular , Intermittent Claudication/genetics , Intermittent Claudication/metabolism , Intermittent Claudication/therapy , Ischemia/diagnosis , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Leg Ulcer/genetics , Leg Ulcer/metabolism , Leg Ulcer/therapy , Male , Middle Aged , Pain Measurement , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology , Prospective Studies , Recovery of Function , Time Factors , Treatment Outcome , Wound Healing , Young Adult
6.
N Engl J Med ; 364(5): 432-42, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21288095

ABSTRACT

BACKGROUND: Arterial calcifications are associated with increased cardiovascular risk, but the genetic basis of this association is unclear. METHODS: We performed clinical, radiographic, and genetic studies in three families with symptomatic arterial calcifications. Single-nucleotide-polymorphism analysis, targeted gene sequencing, quantitative polymerase-chain-reaction assays, Western blotting, enzyme measurements, transduction rescue experiments, and in vitro calcification assays were performed. RESULTS: We identified nine persons with calcifications of the lower-extremity arteries and hand and foot joint capsules: all five siblings in one family, three siblings in another, and one patient in a third family. Serum calcium, phosphate, and vitamin D levels were normal. Affected members of Family 1 shared a single 22.4-Mb region of homozygosity on chromosome 6 and had a homozygous nonsense mutation (c.662C→A, p.S221X) in NT5E, encoding CD73, which converts AMP to adenosine. Affected members of Family 2 had a homozygous missense mutation (c.1073G→A, p.C358Y) in NT5E. The proband of Family 3 was a compound heterozygote for c.662C→A and c.1609dupA (p.V537fsX7). All mutations found in the three families result in nonfunctional CD73. Cultured fibroblasts from affected members of Family 1 showed markedly reduced expression of NT5E messenger RNA, CD73 protein, and enzyme activity, as well as increased alkaline phosphatase levels and accumulated calcium phosphate crystals. Genetic rescue experiments normalized the CD73 and alkaline phosphatase activity in patients' cells, and adenosine treatment reduced the levels of alkaline phosphatase and calcification. CONCLUSIONS: We identified mutations in NT5E in members of three families with symptomatic arterial and joint calcifications. This gene encodes CD73, which converts AMP to adenosine, supporting a role for this metabolic pathway in inhibiting ectopic tissue calcification. (Funded by the National Human Genome Research Institute and the National Heart, Lung, and Blood Institute of the National Institutes of Health.).


Subject(s)
5'-Nucleotidase/genetics , Atherosclerosis/genetics , Calcinosis/genetics , Joint Diseases/genetics , Mutation , 5'-Nucleotidase/metabolism , Arteries/pathology , Chromosomes, Human, Pair 6 , Codon, Nonsense , DNA Mutational Analysis , Female , Fibroblasts/metabolism , Genotype , Humans , Intermittent Claudication/genetics , Lower Extremity/blood supply , Lower Extremity/diagnostic imaging , Mutation, Missense , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Radiography
7.
J Vasc Surg ; 57(5): 1179-85.e1-2, 2013 May.
Article in English | MEDLINE | ID: mdl-23312942

ABSTRACT

OBJECTIVE: Factors responsible for the variability in outcomes after lower extremity vein bypass grafting (LEVBG) are poorly understood. Recent evidence has suggested that a single nucleotide polymorphism (SNP) in the promoter region of the p27(Kip1) gene, a cell-cycle regulator, is associated with coronary in-stent restenosis. We hypothesized an association with vein graft patency. METHODS: This was a retrospective genetic association study nested within a prospective cohort of 204 patients from three referral centers undergoing LEVBG for claudication or critical ischemia. The main outcome measure was primary vein graft patency. RESULTS: All patients were followed up for a minimum of 1 year with duplex graft surveillance (median follow-up, 893 days; interquartile range, 539-1315). Genomic DNA was isolated and SNP analysis for the p27(Kip1)-838C>A variants was performed. Allele frequencies were correlated with graft outcome using survival analysis and Cox proportional hazards modeling. The p27(Kip1)-838C>A allele frequencies observed were CA, 53%; CC, 30%; and AA, 17%, satisfying Hardy-Weinberg equilibrium. Race (P = .025) and history of coronary artery disease (P = .027) were different across the genotypes; all other baseline variables were similar. Primary graft patency was greater among patients with the -838AA genotype (75% AA vs 55% CA/CC at 3 years; P = .029). In a Cox proportional hazards model including age, sex, race, diabetes, critical limb ischemia, redo (vs primary) bypass, vein type, and baseline C-reactive protein level, the p27(Kip1)-838AA genotype was significantly associated with higher graft patency (hazard ratio for failure, 0.4; 95% confidence interval, 0.17-0.93). Genotype was also associated with early (0-1 month) changes in graft lumen diameter by ultrasound imaging. CONCLUSIONS: These data suggest that the p27(Kip1)-838C>A SNP is associated with LEVBG patency and, together with previous reports, underscore a central role for p27(Kip1) in the generic response to vascular injury.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Graft Occlusion, Vascular/genetics , Intermittent Claudication/surgery , Ischemia/surgery , Lower Extremity/blood supply , Peripheral Arterial Disease/surgery , Polymorphism, Single Nucleotide , Vascular Grafting/adverse effects , Vascular Patency/genetics , Veins/transplantation , Aged , Critical Illness , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Graft Occlusion, Vascular/diagnostic imaging , Graft Occlusion, Vascular/physiopathology , Humans , Intermittent Claudication/genetics , Intermittent Claudication/physiopathology , Ischemia/genetics , Ischemia/physiopathology , Male , Middle Aged , Multivariate Analysis , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/physiopathology , Phenotype , Promoter Regions, Genetic , Proportional Hazards Models , Retrospective Studies , Risk Factors , Time Factors , Treatment Outcome , Ultrasonography, Doppler, Duplex , United States , Veins/diagnostic imaging , Veins/physiopathology
8.
Circulation ; 124(16): 1765-73, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21947297

ABSTRACT

BACKGROUND: Hypoxia-inducible factor-1α (HIF-1α) is a transcriptional regulatory factor that orchestrates cellular responses to hypoxia. It increases collateral vessel growth and blood flow in models of hind-limb ischemia. This study tested whether intramuscular administration of Ad2/HIF-1α/VP16, an engineered recombinant type 2 adenovirus vector encoding constitutively active HIF-1α, improves walking time in patients with peripheral artery disease and intermittent claudication. METHODS AND RESULTS: Two hundred eighty-nine patients with claudication were randomized in a double-blind manner to 1 of 3 doses of Ad2/HIF-1α/VP16 (2×10(9), 2×10(10), or 2×10(11) viral particles) or placebo, administered by 20 intramuscular injections to each leg. Graded treadmill tests were performed at baseline and then 3, 6, and 12 months after treatment. The primary end point was the change in peak walking time from baseline to 6 months. The secondary end point was change in claudication onset time, and tertiary end points included changes in ankle-brachial index and quality-of-life assessments. Median peak walking time increased by 0.82 minutes (interquartile range, -0.05-1.93 minutes) in the placebo group and by 0.82 minutes (interquartile range, -0.07-2.12 minutes), 0.28 minutes (interquartile range, -0.37-1.70 minutes), and 0.78 minutes (interquartile range, -0.02-2.10 minutes) in the HIF-1α 2×10(9), 2×10(10), and 2×10(11) viral particle groups, respectively (P=NS between placebo and each HIF-1α treatment group). There were no significant differences in claudication onset time, ankle-brachial index, or quality-of-life measurements between the placebo and each HIF-1α group. CONCLUSIONS: Gene therapy with intramuscular administration of Ad2/HIF-1α/VP16 is not an effective treatment for patients with intermittent claudication. Clinical Trial Registration-URL: http://www.clinicaltrials.gov. Unique identifier: NCT00117650.


Subject(s)
Genetic Therapy/methods , Hypoxia-Inducible Factor 1, alpha Subunit/administration & dosage , Intermittent Claudication/therapy , Walking , Adenoviridae/genetics , Adult , Aged , Aged, 80 and over , Double-Blind Method , Exercise Test/drug effects , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , Intermittent Claudication/genetics , Intermittent Claudication/physiopathology , Male , Middle Aged , Peripheral Arterial Disease/therapy , Quality of Life , Treatment Failure
9.
PLoS One ; 17(3): e0265393, 2022.
Article in English | MEDLINE | ID: mdl-35298547

ABSTRACT

BACKGROUND: Intermittent claudication (IC) is a common manifestation of peripheral arterial disease. Some patients with IC experience a rise in Urinary N-acetyl-ß-D-Glucosaminidase (NAG)/ Creatinine (Cr) ratio, a marker of renal injury, following exercise. In this study, we aim to investigate whether peripheral blood mononuclear cells (PBMC) from patients with IC who exhibit a rise in urinary NAG/ Cr ratio following exercise exhibit differential IL-10/ IL-12 ratio and gene expression compared to those who do not have a rise in NAG/ Cr ratio. METHODS: We conducted a single center observational cohort study of patients diagnosed with IC. Blood and urine samples were collected at rest and following a standardised treadmill exercise protocol. For comparative analysis patients were separated into those with any rise in NAG/Cr ratio (Group 1) and those with no rise in NAG/Cr ratio (Group 2) post exercise. Isolated PBMC from pre- and post-exercise blood samples were analysed using flow cytometry. PBMC were also cultured for 20 hours to perform further analysis of IL-10 and IL-12 cytokine levels. RNA-sequencing analysis was performed to identify differentially expressed genes between the groups. RESULTS: 20 patients were recruited (Group 1, n = 8; Group 2, n = 12). We observed a significantly higher IL-10/IL-12 ratio in cell supernatant from participants in Group 1, as compared to Group 2, on exercise at 20 hours incubation; 47.24 (IQR 9.70-65.83) vs 6.13 (4.88-12.24), p = 0.04. 328 genes were significantly differentially expressed between Group 1 and 2. The modulated genes had signatures encompassing hypoxia, metabolic adaptation to starvation, inflammatory activation, renal protection, and oxidative stress. DISCUSSION: Our results suggest that some patients with IC have an altered immune status making them 'vulnerable' to systemic inflammation and renal injury following exercise. We have identified a panel of genes which are differentially expressed in this group of patients.


Subject(s)
Acute Kidney Injury , Intermittent Claudication , Acetylglucosaminidase/urine , Acute Kidney Injury/metabolism , Biomarkers/urine , Creatinine/urine , Cytokines/genetics , Female , Gene Expression , Humans , Interleukin-10/genetics , Interleukin-12/genetics , Intermittent Claudication/genetics , Leukocytes, Mononuclear/metabolism , Male
10.
Am J Physiol Regul Integr Comp Physiol ; 300(3): R595-604, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21209383

ABSTRACT

The arterial blockage in patients with peripheral arterial disease (PAD) restricts oxygen delivery to skeletal muscles distal to the blockage. In advanced-stage PAD patients, this creates a chronic ischemic condition in the affected muscles. However, in the majority of PAD patients, the muscles distal to the blockage only become ischemic during physical activity when the oxygen demands of these muscles are increased. Therefore, the skeletal muscle of most PAD patients undergoes repeated cycles of low-grade ischemia-reperfusion each time the patient is active and then rests. This has been speculated to contribute to the biochemical and morphological myopathies observed in PAD patients. The current study aimed to determine, using a rodent model, whether repeated hind limb muscle contractions during blood flow restriction to the hind limb muscles increases NF-κB activity. We, subsequently, determined whether an increase in NF-κB activity during this condition is required for the increased transcription of specific atrophy-related genes and muscle fiber atrophy. We found that hind limb muscle contractions during blood flow restriction to the limb increased NF-κB activity, the transcription of specific atrophy-related genes, and caused a 35% decrease in muscle fiber cross-sectional area. We further found that inhibition of NF-κB activity, via gene transfer of a dominant-negative inhibitor of κBα (d.n. IκBα), prevented the increase in atrophy gene expression and muscle fiber atrophy. These findings demonstrate that when blood flow to skeletal muscle is restricted, repeated cycles of muscle contraction can cause muscle fiber atrophy that requires NF-κB-IκBα signaling.


Subject(s)
I-kappa B Proteins/metabolism , Intermittent Claudication/metabolism , Ischemia/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Animals , Disease Models, Animal , Electric Stimulation , Electroporation , Gene Expression Regulation , Gene Transfer Techniques , Hindlimb , I-kappa B Proteins/genetics , Intermittent Claudication/genetics , Intermittent Claudication/pathology , Intermittent Claudication/physiopathology , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Ligation , Male , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Regional Blood Flow , Time Factors , Transcription, Genetic
11.
J Vasc Surg ; 51(4): 1003-5, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20080008

ABSTRACT

Nonatherosclerotic etiologies of arterial insufficiency are uncommon but important causes of chronic lower extremity ischemia. We report a patient with multiple hereditary exostoses (MHE) presenting with lifestyle-limiting lower extremity claudication and popliteal artery occlusion secondary to a large osteochondroma. The presence of MHE with associated osteochondroma resulting in arterial occlusion is a rare condition. Management strategies for treating large osteochondromas adjacent to or with vessel involvement in asymptomatic patients remain undefined.


Subject(s)
Arterial Occlusive Diseases/genetics , Exostoses, Multiple Hereditary/genetics , Intermittent Claudication/genetics , Ischemia/genetics , Lower Extremity/blood supply , Popliteal Artery , Tibia/pathology , Angiography, Digital Subtraction , Arterial Occlusive Diseases/diagnosis , Arterial Occlusive Diseases/surgery , Constriction, Pathologic , Exostoses, Multiple Hereditary/complications , Exostoses, Multiple Hereditary/pathology , Female , Humans , Intermittent Claudication/diagnosis , Intermittent Claudication/surgery , Ischemia/diagnosis , Ischemia/surgery , Middle Aged , Popliteal Artery/diagnostic imaging , Popliteal Artery/surgery , Saphenous Vein/transplantation , Tomography, X-Ray Computed , Treatment Outcome , Ultrasonography, Doppler, Duplex , Vascular Surgical Procedures
12.
Cardiovasc Res ; 116(7): 1386-1397, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31504257

ABSTRACT

AIMS: MicroRNA-378a, highly expressed in skeletal muscles, was demonstrated to affect myoblasts differentiation and to promote tumour angiogenesis. We hypothesized that miR-378a could play a pro-angiogenic role in skeletal muscle and may be involved in regeneration after ischaemic injury in mice. METHODS AND RESULTS: Silencing of miR-378a in murine C2C12 myoblasts did not affect differentiation but impaired their secretory angiogenic potential towards endothelial cells. miR-378a knockout (miR-378a-/-) in mice resulted in a decreased number of CD31-positive blood vessels and arterioles in gastrocnemius muscle. In addition, diminished endothelial sprouting from miR-378a-/- aortic rings was shown. Interestingly, although fibroblast growth factor 1 (Fgf1) expression was decreased in miR-378a-/- muscles, this growth factor did not mediate the angiogenic effects exerted by miR-378a. In vivo, miR-378a knockout did not affect the revascularization of the ischaemic muscles in both normo- and hyperglycaemic mice subjected to femoral artery ligation (FAL). No difference in regenerating muscle fibres was detected between miR-378a-/- and miR-378+/+ mice. miR-378a expression temporarily declined in ischaemic skeletal muscles of miR-378+/+ mice already on Day 3 after FAL. At the same time, in the plasma, the level of miR-378a-3p was enhanced. Similar elevation of miR-378a-3p was reported in the plasma of patients with intermittent claudication in comparison to healthy donors. Local adeno-associated viral vectors-based miR-378a overexpression was enough to improve the revascularization of the ischaemic limb of wild-type mice on Day 7 after FAL, what was not reported after systemic delivery of vectors. In addition, the number of infiltrating CD45+ cells and macrophages (CD45+ CD11b+ F4/80+ Ly6G-) was higher in the ischaemic muscles of miR-378a-/- mice, suggesting an anti-inflammatory action of miR-378a. CONCLUSIONS: Data indicate miR-378a role in the pro-angiogenic effect of myoblasts and vascularization of skeletal muscle. After the ischaemic insult, the anti-angiogenic effect of miR-378a deficiency might be compensated by enhanced inflammation.


Subject(s)
Ischemia/metabolism , MicroRNAs/metabolism , Muscle, Skeletal/blood supply , Myoblasts, Skeletal/metabolism , Neovascularization, Physiologic , Regeneration , Aged , Animals , Case-Control Studies , Cell Line , Disease Models, Animal , Female , Genetic Therapy , Humans , Intermittent Claudication/blood , Intermittent Claudication/genetics , Ischemia/genetics , Ischemia/physiopathology , Ischemia/therapy , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged
14.
Heart Vessels ; 24(5): 321-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19784813

ABSTRACT

We aim to determine the efficacy and safety of gene and cell angiogenic therapies in the treatment of peripheral arterial disease (PAD) and evaluate them for the first time by a meta-analysis. We include in the formal meta-analysis only the randomized placebo-controlled phase 2 studies with any angiogenic gene or cell therapy modality to treat patients with PAD (intermittent claudication, ulcer or critical ischemia) identified by electronic search in MEDLINE and EMBASE databases (1980 to date). Altogether, 543 patients are analyzed from six randomized, controlled trials that are comparable with regard to patient selection, study design, and endpoints. We perform the meta-analysis regarding clinical improvement (improvement of peak walk time, relief in rest pain, ulcer healing or limb salvage) and rate of adverse events. At the end of treatment, therapeutic angiogenesis shows a significantly clinical improvement as compared to placebo in patients with PAD (odds ratio [OR] = 1.437; 95% confidence interval [CI] = 1.03-2.00; P = 0.033). The response rate (improvement of peak walk time) of the pooled groups according to clinical severity does not significantly differ for gene therapy as compared with placebo in the treatment of claudicating patients (OR = 1.304; 95% CI = 0.90-1.89; P = 0.16). Otherwise, we find significant efficacy of the treatment in critical ischemia (OR = 2.20; 95% CI = 1.01-4.79; P = 0.046). The adverse events rates show a slightly significantly higher risk of potential nonserious adverse events (edema, hypotension, proteinuria) in the treated group (OR = 1.81; 95% CI = 1.01-3.38; P = 0.045). We find no differences in mortality from any cause, malignancy, or retinopathy. The patients with PAD, and particularly those with critical ischemia, improve their symptoms when treated with angiogenic gene and cell therapy with acceptable tolerability.


Subject(s)
Cell Transplantation , Genetic Therapy , Intermittent Claudication/therapy , Ischemia/therapy , Neovascularization, Physiologic/genetics , Peripheral Vascular Diseases/therapy , Cell Transplantation/adverse effects , Clinical Trials, Phase II as Topic , Double-Blind Method , Genetic Therapy/adverse effects , Humans , Intermittent Claudication/etiology , Intermittent Claudication/genetics , Intermittent Claudication/physiopathology , Ischemia/etiology , Ischemia/genetics , Ischemia/physiopathology , Odds Ratio , Pain/etiology , Pain/prevention & control , Pain Measurement , Peripheral Vascular Diseases/complications , Peripheral Vascular Diseases/genetics , Peripheral Vascular Diseases/physiopathology , Randomized Controlled Trials as Topic , Recovery of Function , Risk Assessment , Risk Factors , Severity of Illness Index , Treatment Outcome , Walking , Wound Healing
15.
J Vasc Surg ; 48(6): 1497-503, 1503.e1, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18829228

ABSTRACT

OBJECTIVE: Studies report clustering of cardiovascular risk factors and increased cardiovascular events in healthy first-degree relatives (FDR) of subjects with intermittent claudication (IC). Family history is an independent risk factor in coronary artery disease but the role of genetic factors is undefined in peripheral arterial disease. The fibrin clot is the final product of the atherothrombotic process and is subject to genetic influence. We proposed that healthy male FDR of subjects with IC possess abnormalities in their fibrin clots. METHODS: This was a case-control family study. The FDR were recruited from claudicants attending vascular surgery out-patient clinics with the control subjects being recruited from the local primary care register. A total of 106 white European male FDR of male subjects with IC were age matched with 107 white European male control subjects from an identical geographic area. The control subjects had no FDR with a history of symptomatic cardiovascular disease, and subjects from both groups were free from a personal history of symptomatic cardiovascular disease or diabetes mellitus. Ex vivo assays for fibrin clot permeation, fiber thickness, factor XIII cross-linking activity, and fibrinolysis were performed on the plasma of the above subjects. In addition, linear regression analysis was undertaken to determine factors associated with clot parameters. RESULTS: For controls and FDR, respectively, fiber thickness by turbidity was 0.75 (0.67-0.93) vs 0.86 (0.75-0.98) (P < .001), and FXIII cross-linking activity was 105% (87-141) vs 133% (103-155) (P < .001). On confocal microscopy, fibers measured 315.8 (307.0-324.6) vs 405.1 (397.6-412.6) nm (P < .001), and lysis front velocity was 12.66 (6.38-18.94) vs 4.83 (2.50-7.17), mum/min (P = .018). Linear regression analysis revealed cholesterol was associated with changes in certain clot parameters. CONCLUSION: The healthy FDR of subjects with IC produce clots which have thicker fibers, increased cross-linking, and resistance to fibrinolysis when compared to controls. This supports the potential genetic basis of peripheral arterial disease and highlights that cholesterol may contribute to this abnormal structure. This suggests that the FDR of subjects with IC, an apparently healthy sub-group of the population, have an elevated cardiovascular risk associated with abnormalities in their clot structure.


Subject(s)
Blood Coagulation/physiology , Family , Fibrin/analysis , Fibrin/physiology , Genetic Predisposition to Disease , Intermittent Claudication/blood , Intermittent Claudication/genetics , Adult , Aged , Case-Control Studies , Genotype , Humans , Male , Microscopy, Confocal , Middle Aged , Polymorphism, Genetic , Prognosis , Risk Factors
16.
Eur J Vasc Endovasc Surg ; 36(2): 132-137, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18396070

ABSTRACT

OBJECTIVES: To investigate the relationship between the Pl(A1/A2) polymorphism and platelet activation and aggregation in patients with Peripheral Arterial Disease (PAD). DESIGN: A prospective single-centre cohort study. METHODS: 45 patients with PAD on aspirin 75mg were recruited and phenotyped/genotyped for the Gp IIb/IIIa Pl(A1/A2) polymorphism. Platelet-Monocyte Aggregation (PMAs) was evaluated using flow-cytometry. RESULTS: The formation of PMAs in the Pl(A2) group was higher but not statistically significant (p=0.17). However, when males were analysed separately, the formation of PMAs was significantly higher in the Pl(A2) group (p=0.0192). No difference was seen in the females. CONCLUSIONS: In this study we show that the Pl(A1/A2) polymorphism primarily affects the aggregation of platelets to monocytes in males. The effect is not observed in females and understanding the mechanism behind this may help elucidate the way the polymorphism alters platelet function in the presence of aspirin.


Subject(s)
Blood Platelets/metabolism , Intermittent Claudication/genetics , Monocytes/metabolism , Peripheral Vascular Diseases/genetics , Platelet Adhesiveness/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Polymorphism, Restriction Fragment Length , Aged , Aspirin/therapeutic use , Blood Platelets/drug effects , Female , Flow Cytometry , Gene Frequency , Genotype , Humans , Intermittent Claudication/blood , Intermittent Claudication/drug therapy , Male , Peripheral Vascular Diseases/blood , Peripheral Vascular Diseases/complications , Peripheral Vascular Diseases/drug therapy , Phenotype , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Prospective Studies , Sex Factors
17.
JCI Insight ; 3(21)2018 11 02.
Article in English | MEDLINE | ID: mdl-30385731

ABSTRACT

The most severe manifestation of peripheral arterial disease (PAD) is critical limb ischemia (CLI). CLI patients suffer high rates of amputation and mortality; accordingly, there remains a clear need both to better understand CLI and to develop more effective treatments. Gastrocnemius muscle was obtained from 32 older (51-84 years) non-PAD controls, 27 claudicating PAD patients (ankle-brachial index [ABI] 0.65 ± 0.21 SD), and 19 CLI patients (ABI 0.35 ± 0.30 SD) for whole transcriptome sequencing and comprehensive mitochondrial phenotyping. Comparable permeabilized myofiber mitochondrial function was paralleled by both similar mitochondrial content and related mRNA expression profiles in non-PAD control and claudicating patient tissues. Tissues from CLI patients, despite being histologically intact and harboring equivalent mitochondrial content, presented a unique bioenergetic signature. This signature was defined by deficits in permeabilized myofiber mitochondrial function and a unique pattern of both nuclear and mitochondrial encoded gene suppression. Moreover, isolated muscle progenitor cells retained both mitochondrial functional deficits and gene suppression observed in the tissue. These findings indicate that muscle tissues from claudicating patients and non-PAD controls were similar in both their bioenergetics profile and mitochondrial phenotypes. In contrast, CLI patient limb skeletal muscles harbor a unique skeletal muscle mitochondriopathy that represents a potentially novel therapeutic site for intervention.


Subject(s)
Intermittent Claudication/genetics , Ischemia/pathology , Mitochondria, Muscle/pathology , Peripheral Arterial Disease/genetics , Aged , Aged, 80 and over , Ankle Brachial Index/methods , Atherosclerosis , Cellular Microenvironment/physiology , Cross-Sectional Studies , Female , Humans , Intermittent Claudication/diagnosis , Intermittent Claudication/physiopathology , Male , Middle Aged , Mitochondria, Muscle/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Peripheral Arterial Disease/complications , Phenotype , RNA, Messenger/genetics , Exome Sequencing/methods
18.
Ther Adv Cardiovasc Dis ; 12(9): 237-246, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29996720

ABSTRACT

BACKGROUND: The effective treatment of chronic lower limb ischemia is one of the most challenging issues confronting vascular surgeons. Current pharmacological therapies play an auxiliary role and cannot prevent disease progression, and new treatment methods are needed. In 2011, a plasmid VEGF65-gene therapy drug was approved in Russia for the treatment of chronic lower limb ischemia ( ClinicalTrials.gov identifier: NCT03068585). The objective of this follow-up study was to evaluate the long-term safety and efficacy of gene therapy in patients with limb ischemia of atherosclerotic genesis. AIMS: To evaluate the long-term safety and efficacy of the therapeutic angiogenesis, 36 patients in the treatment group (pl- VEGF165) and 12 patients in the control group participated in a 5-year follow-up study. Planned examinations were carried out annually for 5 years after pl- VEGF165 administration. RESULTS: Differences in the frequency of major cardiovascular events (pl- VEGF165 5/36 versus control 2/12; p = 0.85), malignancies (pl- VEGF165 1/36 versus control 0/12; p = 0.38) and impaired vision (there was none in either group) over the 5-year follow-up period did not achieve statistical significance. The target limb salvage was 95% ( n = 36) and 67% ( n = 12) in the pl- VEGF165 and control groups, respectively. The pain-free walking distance value increased by 288% from 105.7 ± 16.5 m to 384 ± 39 m in the treatment group by the end of the fifth year, with a peak of 410.6 ± 86.1 m achieved by the end of the third year. The ankle-brachial index (ABI) increased from 0.47 ± 0.01 to 0.56 ± 0.02 by the end of the first year, with a subsequent slight decrease to 0.51 ± 0.02 by the fifth year. The maximum increment of transcutaneous oximetry test (tcoO2) by 36%, from 66.6 ± 3.7 mm Hg to 90.7 ± 4.9 mm Hg, was observed by the end of the second year. CONCLUSION: The therapeutic effect of angiogenesis induction by gene therapy persists for 5 years.


Subject(s)
Genetic Therapy/methods , Intermittent Claudication/therapy , Ischemia/therapy , Lower Extremity/blood supply , Peripheral Arterial Disease/therapy , Vascular Endothelial Growth Factor A/genetics , Aged , Ankle Brachial Index , Blood Gas Monitoring, Transcutaneous , Exercise Tolerance , Female , Follow-Up Studies , Genetic Therapy/adverse effects , Humans , Intermittent Claudication/genetics , Intermittent Claudication/metabolism , Intermittent Claudication/physiopathology , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Limb Salvage , Male , Middle Aged , Neovascularization, Physiologic , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology , Recovery of Function , Russia , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/biosynthesis , Walk Test
19.
Mech Ageing Dev ; 127(3): 307-13, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16405962

ABSTRACT

Mutations in the WRN gene lead to the Werner syndrome (WS), which resembles premature aging. Here, we hypothesize that genetic variations in the WRN gene may also influence aging-trajectories in the population at large. To test this hypothesis, we assessed the impact of the i1-C/T, L1074F and C1367R polymorphisms in the WRN gene on the occurrence of cardiovascular pathologies, on cognitive performance and on the risks of all-cause, cardiovascular and cancer mortalities in the population-based Leiden 85-plus Study. This prospective follow-up study includes 1,245 participants aged 85 years and older, with a total follow-up of 5,164 person-years. At baseline the risks of myocardial infarction, myocardial ischemia, intermittent claudication, arterial surgery and stroke dependent on the i1-C/T, L1074F and C1367R polymorphisms, did not vary between the different genotypes. Also no differences in cognitive functioning were observed, except for attention, where carriers of the 1367R allele performed worse compared to the 1367C homozygotes (94.2 (4.35) versus 84.8 (1.84), p=0.04). Mortality risks, calculated separately for all SNPs, were similar between the different genotype carriers of the i1-C/T, L1074F and C1367R polymorphisms, showing no evidence of altered survival. In conclusion, the i1-C/T, L1074F and C1367R polymorphisms in the WRN gene do not influence the aging-trajectories and survival in the population at large.


Subject(s)
Aging/genetics , Aging/pathology , Amino Acid Substitution , DNA Helicases/genetics , Point Mutation , Polymorphism, Single Nucleotide , Adult , Aged, 80 and over , Cognition Disorders/genetics , Cognition Disorders/mortality , Cognition Disorders/pathology , Exodeoxyribonucleases , Female , Follow-Up Studies , Heterozygote , Homozygote , Humans , Intermittent Claudication/genetics , Intermittent Claudication/mortality , Intermittent Claudication/pathology , Male , Myocardial Infarction/genetics , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Prospective Studies , RecQ Helicases , Werner Syndrome Helicase
20.
Atherosclerosis ; 183(2): 349-54, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15899485

ABSTRACT

Human serum paraoxonase-1 (PON1) is thought to play a role in the favorable vascular effects of high-density lipoproteins, mainly through a reduction in low-density lipoprotein oxidation. Endothelial dysfunction, characterized by an impaired capacity of the arteries to dilate in response to a number of stimuli, represents the earliest stage of atherosclerosis. We performed the present study in 37 patients with peripheral arterial disease, with the aim of investigating the influence of PON1 Q192R polymorphism and activity on peripheral endothelial function, evaluated as brachial-artery flow-mediated vasodilation (FMV). Patients with the R allele (QR or RR genotype, n=19) had significantly higher PON1 activity [408 U/mL (309-456) versus 180 U/mL (141-243), p<0.001] and greater brachial FMV (5.7+/-3.9% versus 3.0+/-2.8%, p<0.001) than those with Q allele (QQ genotype, n=18). In the whole population, PON1 activity showed a direct relation to brachial FMV (r=0.46, p=0.004). In a multivariate linear regression analysis, the only independent predictors of brachial FMV were PON1 activity (beta=0.40, p=0.008), brachial-artery diameter (beta=-0.39, p=0.01) and male sex (beta=-0.27, p=0.04). These finding support the importance of PON1 activity as a modulating factor of the endothelial function.


Subject(s)
Aryldialkylphosphatase/genetics , DNA/genetics , Endothelium, Vascular/physiopathology , Intermittent Claudication/blood , Polymorphism, Genetic , Aged , Alleles , Aryldialkylphosphatase/blood , Blood Pressure , Brachial Artery/diagnostic imaging , Brachial Artery/physiology , Female , Follow-Up Studies , Genotype , Humans , Intermittent Claudication/genetics , Intermittent Claudication/physiopathology , Lipoproteins, HDL/blood , Male , Polymerase Chain Reaction , Prognosis , Sex Factors , Ultrasonography , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL