1
|
Zalewski D, Chmiel P, Kołodziej P, Kocki M, Feldo M, Kocki J, Bogucka-Kocka A. Key Regulators of Angiogenesis and Inflammation Are Dysregulated in Patients with Varicose Veins. Int J Mol Sci 2024; 25:6785. [PMID: 38928491 PMCID: PMC11204110 DOI: 10.3390/ijms25126785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Varicose veins (VVs) are the most common manifestation of chronic venous disease (CVD) and appear as abnormally enlarged and tortuous superficial veins. VVs result from functional abnormalities in the venous circulation of the lower extremities, such as venous hypertension, venous valve incompetence, and venous reflux. Previous studies indicate that enhanced angiogenesis and inflammation contribute to the progression and onset of VVs; however, dysregulations in signaling pathways associated with these processes in VVs patients are poorly understood. Therefore, in our study, we aimed to identify key regulators of angiogenesis and inflammation that are dysregulated in patients with VVs. Expression levels of 18 genes were analyzed in peripheral blood mononuclear cells (PBMC) using real-time PCR, as well as plasma levels of 6 proteins were investigated using ELISA. Higher levels of CCL5, PDGFA, VEGFC, TGF-alpha, TGF-beta 1, and VEGF-A, as well as lower levels of VEGFB and VEGF-C, were found to be statistically significant in the VV group compared to the control subjects without VVs. None of the analyzed factors was associated with the venous localization of the varicosities. The presented study identified dysregulations in key angiogenesis- and inflammation-related factors in PBMC and plasma from VVs patients, providing new insight into molecular mechanisms that could contribute to the development of VVs and point out promising candidates for circulatory biomarkers of this disease.
Collapse
Affiliation(s)
- Daniel Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (A.B.-K.)
| | - Paulina Chmiel
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (A.B.-K.)
| | - Przemysław Kołodziej
- Laboratory of Diagnostic Parasitology, Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Marcin Kocki
- Department of Neonatology and Neonatal Intensive Care, Independent Public Hospital No. 4 in Lublin, 8 Jaczewski St., 20-954 Lublin, Poland;
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland;
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland;
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (A.B.-K.)
| |
Collapse
|
2
|
Nadasy GL, Patai BB, Molnar AA, Hetthessy JR, Tokes AM, Varady Z, Dornyei G. Vicious Circle With Venous Hypertension, Irregular Flow, Pathological Venous Wall Remodeling, and Valve Destruction in Chronic Venous Disease: A Review. Angiology 2024:33197241256680. [PMID: 38839285 DOI: 10.1177/00033197241256680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Substantial advances occurred in phlebological practice in the last two decades. With the use of modern diagnostic equipment, the patients' venous hemodynamics can be examined in detail in everyday practice. Application of venous segments for arterial bypasses motivated studies on the effect of hemodynamic load on the venous wall. New animal models have been developed to study hemodynamic effects on the venous system. In vivo and in vitro studies revealed cellular phase transitions of venous endothelial, smooth muscle, and fibroblastic cells and changes in connective tissue composition, under hemodynamic load and at different locations of the chronically diseased venous system. This review is an attempt to integrate our knowledge from epidemiology, paleoanthropology and anthropology, clinical and experimental hemodynamic studies, histology, cell physiology, cell pathology, and molecular biology on the complex pathomechanism of this frequent disease. Our conclusion is that the disease is initiated by limited genetic adaptation of mankind not to bipedalism but to bipedalism in the unmoving standing or sitting position. In the course of the disease several pathologic vicious circles emerge, sustained venous hypertension inducing cellular phase transitions, chronic wall inflammation, apoptosis of cells, pathologic dilation, and valvular damage which, in turn, further aggravate the venous hypertension.
Collapse
Affiliation(s)
- Gyorgy L Nadasy
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Andrea A Molnar
- Department of Cardiology, Semmelweis University, Budapest, Hungary
| | | | - Anna-Maria Tokes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | | | - Gabriella Dornyei
- Department of Morphology and Physiology, Health Science Faculty, Semmelweis University, Budapest, Hungary
| |
Collapse
|
3
|
Guo P, Fang Q, Wang Y. Associations between varicose veins and heart failure: A genetic correlation and mendelian randomization study. Medicine (Baltimore) 2024; 103:e38175. [PMID: 38758877 PMCID: PMC11098184 DOI: 10.1097/md.0000000000038175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/17/2024] [Indexed: 05/19/2024] Open
Abstract
Varicose veins and heart failure (HF) are increasingly prevalent. Although numbers of observational studies have indicated that varicose veins might contribute to the risk of HF, the causal relationship between them remains unclear due to the uncontrolled confounding factors and reverse causation bias. Therefore, this study aimed to explore the potential causal relationship between varicose veins and HF. Based on publicly released genome-wide association studies (GWAS), gene correlation was assessed using linkage disequilibrium score (LDSC) regression, and we conducted a two-sample Mendelian randomization (TSMR) analysis to infer the causal relationship. We performed the Inverse variance weighted (IVW) method as the primary analysis, and used Weighted median, MR-Egger, weighted mode, simple mode, and MR-pleiotropy residual sum and outlier (MR-PRESSO) methods to detect and correct for horizontal pleiotropy. LDSC revealed there was a positive genetic correlation between varicose veins and HF (rg = 0.1726184, Se = 0.04511803, P = .0001). The results of the IVW method indicated that genetically predicted varicose veins were associated with an increased risk of HF (odds ratio (OR) = 1.03; 95% confidence interval (CI): 1.01-1.06; P = .009). Our findings illustrated the significant causal effect of varicose veins on HF, suggesting that people with varicose veins might have a higher risk of HF. The results provided a novel and important perspective into the development mechanism of HF.
Collapse
Affiliation(s)
- Ping Guo
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Qin Fang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yan Wang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
4
|
Gwozdzinski L, Pieniazek A, Gwozdzinski K. Factors Influencing Venous Remodeling in the Development of Varicose Veins of the Lower Limbs. Int J Mol Sci 2024; 25:1560. [PMID: 38338837 PMCID: PMC10855638 DOI: 10.3390/ijms25031560] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
One of the early symptoms of chronic venous disease (CVD) is varicose veins (VV) of the lower limbs. There are many etiological environmental factors influencing the development of chronic venous insufficiency (CVI), although genetic factors and family history of the disease play a key role. All these factors induce changes in the hemodynamic in the venous system of the lower limbs leading to blood stasis, hypoxia, inflammation, oxidative stress, proteolytic activity of matrix metalloproteinases (MMPs), changes in microcirculation and, consequently, the remodeling of the venous wall. The aim of this review is to present current knowledge on CVD, including the pathophysiology and mechanisms related to vein wall remodeling. Particular emphasis has been placed on describing the role of inflammation and oxidative stress and the involvement of extracellular hemoglobin as pathogenetic factors of VV. Additionally, active substances used in the treatment of VV were discussed.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland;
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| |
Collapse
|
5
|
He C, Ye P, Zhang X, Esmaeili E, Li Y, Lü P, Cai C. The Role of TGF-β Signaling in Saphenous Vein Graft Failure after Peripheral Arterial Disease Bypass Surgery. Int J Mol Sci 2023; 24:10381. [PMID: 37373529 DOI: 10.3390/ijms241210381] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Saphenous vein bypass grafting is an effective technique used to treat peripheral arterial disease (PAD). However, restenosis is the major clinical challenge for the graft vessel among people with PAD postoperation. We hypothesize that there is a common culprit behind arterial occlusion and graft restenosis. To investigate this hypothesis, we found TGF-β, a gene specifically upregulated in PAD arteries, by bioinformatics analysis. TGF-β has a wide range of biological activities and plays an important role in vascular remodeling. We discuss the molecular pathway of TGF-β and elucidate its mechanism in vascular remodeling and intimal hyperplasia, including EMT, extracellular matrix deposition, and fibrosis, which are the important pathways contributing to stenosis. Additionally, we present a case report of a patient with graft restenosis linked to the TGF-β pathway. Finally, we discuss the potential applications of targeting the TGF-β pathway in the clinic to improve the long-term patency of vein grafts.
Collapse
Affiliation(s)
- Changhuai He
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pin Ye
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xuecheng Zhang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Elham Esmaeili
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ping Lü
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
6
|
Reddy TP, Glynn SA, Billiar TR, Wink DA, Chang JC. Targeting Nitric Oxide: Say NO to Metastasis. Clin Cancer Res 2023; 29:1855-1868. [PMID: 36520504 PMCID: PMC10183809 DOI: 10.1158/1078-0432.ccr-22-2791] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/24/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022]
Abstract
Utilizing targeted therapies capable of reducing cancer metastasis, targeting chemoresistant and self-renewing cancer stem cells, and augmenting the efficacy of systemic chemo/radiotherapies is vital to minimize cancer-associated mortality. Targeting nitric oxide synthase (NOS), a protein within the tumor microenvironment, has gained interest as a promising therapeutic strategy to reduce metastatic capacity and augment the efficacy of chemo/radiotherapies in various solid malignancies. Our review highlights the influence of nitric oxide (NO) in tumor progression and cancer metastasis, as well as promising preclinical studies that evaluated NOS inhibitors as anticancer therapies. Lastly, we highlight the prospects and outstanding challenges of using NOS inhibitors in the clinical setting.
Collapse
Affiliation(s)
- Tejaswini P. Reddy
- Texas A&M University Health Science Center, Bryan, Texas
- Houston Methodist Research Institute, Houston, Texas
- Houston Methodist Neal Cancer Center, Houston, Texas
| | - Sharon A. Glynn
- Prostate Cancer Institute, National University of Ireland Galway, Galway, Ireland
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David A. Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, Maryland
| | - Jenny C. Chang
- Houston Methodist Research Institute, Houston, Texas
- Houston Methodist Neal Cancer Center, Houston, Texas
| |
Collapse
|
7
|
Hemodynamic and neurobiological factors for the development of chronic pelvic pain in patients with pelvic venous disorder. J Vasc Surg Venous Lymphat Disord 2023; 11:610-618.e3. [PMID: 36781107 DOI: 10.1016/j.jvsv.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/18/2022] [Accepted: 01/05/2023] [Indexed: 02/13/2023]
Abstract
OBJECTIVE The study was aimed at the identification of hemodynamic and neurobiological factors for the development of chronic pelvic pain (CPP) in patients with pelvic venous disorder (PeVD) using ultrasound, radionuclide, and enzyme immunoassay methods. METHODS This cohort study included 110 consecutive patients with PeVD and 20 healthy controls. Seventy patients with PeVD had symptoms (CPP in 100% of cases, discomfort in hypogastrium, dyspareunia, vulvar varices, and dysuria), and 40 were asymptomatic. Patients underwent clinical examination, duplex ultrasound study of the pelvic veins and lower extremities, and single-photon emission computed tomography of the pelvic veins with in vivo labeled red blood cells. The prevalence, duration, severity, and pattern of reflux in the pelvic veins, as well as the severity of pelvic venous congestion, were evaluated. Healthy controls underwent only clinical and duplex ultrasound examination. All 130 patients were assessed using enzyme immunoassays to determine plasma levels of calcitonin gene-related peptide (CGRP) and substance P (SP). RESULTS Symptomatic patients with PeVD had a higher prevalence of reflux in the ovarian veins (OVs) than asymptomatic ones (45.7% vs 10%, respectively; P = .001) and a greater reflux duration (4.1 ± 1.7 seconds vs 1.4 ± 0.3 seconds; P = .002), although no differences in the OV diameter were found. Similar results were obtained when comparing the diameters of the parametrial veins (PVs) and the duration of reflux in them. Type II/III reflux (greater than 2 seconds) was identified in 41.4% of symptomatic and in only 5% of asymptomatic patients (P = .001). Among patients with CPP, 24.2% had a combined reflux in the OVs, PVs, and uterine veins, and 45.7% had a combined reflux in the OVs and PVs, whereas 90% of patients without CPP had only an isolated reflux in the PVs. The pelvic venous congestion was moderate or severe in 95.7% of patients with CPP and in only 15% patients without CPP (P = .001). In patients with PeVD, the presence of CPP was associated with higher levels of CGRP and SP compared with asymptomatic patients (CGRP: 0.48 ± 0.06 vs 0.19 ± 0.02 ng/mL, respectively, P = .001; SP: 0.38 ± 0.08 vs 0.13 ± 0.03 ng/mL, P = .001). CONCLUSIONS In patients with PeVD, significant hemodynamic and neurobiological factors for the CPP development were found to be reflux in the pelvic veins greater than 2 seconds, involvement of several venous collectors, and increased plasma levels of CGRP and SP.
Collapse
|
8
|
Helkkula P, Hassan S, Saarentaus E, Vartiainen E, Ruotsalainen S, Leinonen JT, Palotie A, Karjalainen J, Kurki M, Ripatti S, Tukiainen T. Genome-wide association study of varicose veins identifies a protective missense variant in GJD3 enriched in the Finnish population. Commun Biol 2023; 6:71. [PMID: 36653477 PMCID: PMC9849365 DOI: 10.1038/s42003-022-04285-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 11/21/2022] [Indexed: 01/19/2023] Open
Abstract
Varicose veins is the most common manifestation of chronic venous disease that displays female-biased incidence. To identify protein-inactivating variants that could guide identification of drug target genes for varicose veins and genetic evidence for the disease prevalence difference between the sexes, we conducted a genome-wide association study of varicose veins in Finns using the FinnGen dataset with 17,027 cases and 190,028 controls. We identified 50 associated genetic loci (P < 5.0 × 10-8) of which 29 were novel including one near ERG with female-specificity (rs2836405-G, OR[95% CI] = 1.09[1.05-1.13], P = 3.1 × 10-8). These also include two X-chromosomal (ARHGAP6 and SRPX) and two autosomal novel loci (TGFB2 and GJD3) with protein-coding lead variants enriched above 56-fold in Finns over non-Finnish non-Estonian Europeans. A low-frequency missense variant in GJD3 (p.Pro59Thr) is exclusively associated with a lower risk for varicose veins (OR = 0.62 [0.55-0.70], P = 1.0 × 10-14) in a phenome-wide scan of the FinnGen data. The absence of observed pleiotropy and its membership of the connexin gene family underlines GJD3 as a potential connexin-modulating therapeutic strategy for varicose veins. Our results provide insights into varicose veins etiopathology and highlight the power of isolated populations, including Finns, to discover genetic variants that inform therapeutic development.
Collapse
Grants
- MC_PC_17228 Medical Research Council
- Academy of Finland (Suomen Akatemia)
- Sydäntutkimussäätiö (Finnish Foundation for Cardiovascular Research)
- Academy of Finland Center of Excellence in Complex Disease Genetics (Grant No 312062), Sigrid Juselius Foundation (S.Ri. and T.T.), University of Helsinki HiLIFE Fellow and Grand Challenge grants (S.Ri.), University of Helsinki three-year research project grant (T.T.), FIMM-EMBL PhD program doctoral funding (S.H.), Nylands Nation, University of Helsinki (P.H.) The FinnGen project is funded by two grants from Business Finland (HUS 4685/31/2016 and UH 4386/31/2016) and the following industry partners: AbbVie Inc., AstraZeneca UK Ltd, Biogen MA Inc., Bristol Myers Squibb (and Celgene Corporation & Celgene International II Sàrl), Genentech Inc., Merck Sharp & Dohme Corp, Pfizer Inc., GlaxoSmithKline Intellectual Property Development Ltd., Sanofi US Services Inc., Maze Therapeutics Inc., Janssen Biotech Inc, Novartis AG, and Boehringer Ingelheim. Following biobanks are acknowledged for delivering biobank samples to FinnGen: Auria Biobank (www.auria.fi/biopankki), THL Biobank (www.thl.fi/biobank), Helsinki Biobank (www.helsinginbiopankki.fi), Biobank Borealis of Northern Finland (https://www.ppshp.fi/Tutkimus-ja-opetus/Biopankki/Pages/Biobank-Borealis-briefly-in-English.aspx), Finnish Clinical Biobank Tampere (www.tays.fi/en-US/Research_and_development/Finnish_Clinical_Biobank_Tampere), Biobank of Eastern Finland (www.ita-suomenbiopankki.fi/en), Central Finland Biobank (www.ksshp.fi/fi-FI/Potilaalle/Biopankki), Finnish Red Cross Blood Service Biobank (www.veripalvelu.fi/verenluovutus/biopankkitoiminta) and Terveystalo Biobank (www.terveystalo.com/fi/Yritystietoa/Terveystalo-Biopankki/Biopankki/).
Collapse
Affiliation(s)
- Pyry Helkkula
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Shabbeer Hassan
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Elmo Saarentaus
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Emilia Vartiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Sanni Ruotsalainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jaakko T Leinonen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Mitja Kurki
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Taru Tukiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
9
|
Avrahami R, Silverberg D, Kolvenbach R, Elias S, Sivak G. Biological autologous excised varicose vein dressing compared to conservative dressing on the ulcer bed during endovenous ablation. Phlebology 2022; 37:386-392. [DOI: 10.1177/02683555221081635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective To compare the use of biological autologous excised varicose vein dressing ( VenoDress) compared to conservative dressing on the ulcer bed during endovenous ablation Methods This retrospective non-blinded study included all consecutive patients with primary or recurrent venous leg ulcers (VLU) with superficial varices treated in one center between September 2019 and October 2020. They all underwent venous ablation, wound debridement, and when needed phlebectomy. On the study group, the excised veins were incised, formed into a sheet, and applied onto the debrided wound bed with the endothelial side facing the wound bed. Adhesion was assessed weekly for 3 weeks. The study group was compared to a control group that underwent similar procedures but with the debrided wound bed treated with low-adherent paraffin dressing. The primary outcome was complete wound healing at 1 and 3 months, and the secondary outcomes were wound-related pain and leg edema. Results Complete wound closure was documented in 17/26 study group patients at 1 month (65%) and in 25/26 (96%) at 3 months. Complete wound closure was documented in 37/82 patients in the control group (45%) and in 67/82 (82%) at 3 months. The 1-month healing rates were significantly in favor of the VenoDress group when adjusted to sex and diabetes: odds ratio = 2.81 (1.05–7.532), p = .04. The preoperative pain level of the study group (as measured by a visual analog scale VAS (0–10) decreased from 4.96 ± 2.71 to 0.73 ± 1.36 at 1 week and that of the control group from 4.8 ± 2 to 1.35 ± 1.38 at 1 week ( p < .001). Conclusion the use of autologous varicose veins as dressing effectively reduced pain in VLU patients compared to conventional techniques. Although its effects on wound closure appear highly promising, further validation is warranted in a randomized comparative study.
Collapse
Affiliation(s)
- Ram Avrahami
- T. L. M. Medical Center, Tel-Aviv, Israel
- Department of Vascular Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Daniel Silverberg
- Department of Vascular Surgery, the Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ralph Kolvenbach
- Department of Vascular Surgery and Endovascular Therapy, SANA Hospital Group, Düsseldorf, Germany
- Academic Teaching Hospital, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Steven Elias
- Center for Vein Disease, Englewood Health Network, Englewood, NJ, USA
| | - Galit Sivak
- T. L. M. Medical Center, Tel-Aviv, Israel
- Department of Vascular Surgery, Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
10
|
Kocarslan S, Kocarslan A, Doganer A, Yasim A. What is the relationship of varicose vein pathogenesis with collagen fibers? Niger J Clin Pract 2022; 25:304-309. [PMID: 35295053 DOI: 10.4103/njcp.njcp_1505_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Aims and Background In this study, the densities of collagen 1 and collagen 4, which are an effective vascular component in the remodelling of varicose veins, were investigated. Materials and Methods The study included primary varicose vein samples of 20 patients and vein samples of 20 healthy controls. Immunohistochemical staining was performed using collagen 1 and collagen 4 antibodies. Histochemical staining was performed using Masson Trichrome. Results In the immunohistochemical analysis of varicose samples, collagen 1 immunostaining was negative in 17 cases (85%) and positive in 3 cases (15%). In healthy venous tissue samples, collagen 1 immunostaining was negative in 12 cases (60%) and positive in 8 cases (40%). There was no statistically significant difference between both groups concerning collagen 1 immunostaining (p > 0.05). In varicose samples, collagen 4 immunostaining was negative in 4 cases (20%) and positive in 16 cases (80%). In healthy venous tissue samples, collagen 4 immunostaining was negative in 13 cases (65%) and positive in 7 cases (35%). Statistical comparison of healthy veins and varicose veins concerning collagen 4 immunostaining showed a significant difference (p = 0.03). In the histochemical analysis of varicose samples, Masson Trichrome staining was negative in 4 cases (20%) and positive in 16 cases (80%). In healthy venous tissue samples, Masson Trichrome staining was negative in 18 cases (90%) and positive in 2 cases (10%). Statistical comparison of healthy veins and varicose veins concerning collagen 4 immunostaining showed a significant difference (p = 0.01). Conclusion The change in the density of collagen types plays an important role in vein wall remodeling.
Collapse
Affiliation(s)
- S Kocarslan
- Department of Pathology, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - A Kocarslan
- Department of Cardiovascular Surgery, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - A Doganer
- Department of Biostatistics, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - A Yasim
- Department of Cardiovascular Surgery, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| |
Collapse
|
11
|
Molnár AÁ, Nádasy GL, Dörnyei G, Patai BB, Delfavero J, Fülöp GÁ, Kirkpatrick AC, Ungvári Z, Merkely B. The aging venous system: from varicosities to vascular cognitive impairment. GeroScience 2021; 43:2761-2784. [PMID: 34762274 PMCID: PMC8602591 DOI: 10.1007/s11357-021-00475-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/12/2021] [Indexed: 10/25/2022] Open
Abstract
Aging-induced pathological alterations of the circulatory system play a critical role in morbidity and mortality of older adults. While the importance of cellular and molecular mechanisms of arterial aging for increased cardiovascular risk in older adults is increasingly appreciated, aging processes of veins are much less studied and understood than those of arteries. In this review, age-related cellular and morphological alterations in the venous system are presented. Similarities and dissimilarities between arterial and venous aging are highlighted, and shared molecular mechanisms of arterial and venous aging are considered. The pathogenesis of venous diseases affecting older adults, including varicose veins, chronic venous insufficiency, and deep vein thrombosis, is discussed, and the potential contribution of venous pathologies to the onset of vascular cognitive impairment and neurodegenerative diseases is emphasized. It is our hope that a greater appreciation of the cellular and molecular processes of vascular aging will stimulate further investigation into strategies aimed at preventing or retarding age-related venous pathologies.
Collapse
Affiliation(s)
- Andrea Ágnes Molnár
- Heart and Vascular Center, Semmelweis University, Városmajor Street 68, 1121, Budapest, Hungary.
| | | | - Gabriella Dörnyei
- Department of Morphology and Physiology, Health Sciences Faculty, Semmelweis University, Budapest, Hungary
| | | | - Jordan Delfavero
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center On Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor Áron Fülöp
- Heart and Vascular Center, Semmelweis University, Városmajor Street 68, 1121, Budapest, Hungary
| | - Angelia C Kirkpatrick
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Veterans Affairs Medical Center, 921 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Zoltán Ungvári
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center On Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Városmajor Street 68, 1121, Budapest, Hungary
| |
Collapse
|
12
|
Spaide RF, Gemmy Cheung CM, Matsumoto H, Kishi S, Boon CJF, van Dijk EHC, Mauget-Faysse M, Behar-Cohen F, Hartnett ME, Sivaprasad S, Iida T, Brown DM, Chhablani J, Maloca PM. Venous overload choroidopathy: A hypothetical framework for central serous chorioretinopathy and allied disorders. Prog Retin Eye Res 2021; 86:100973. [PMID: 34029721 DOI: 10.1016/j.preteyeres.2021.100973] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/11/2021] [Accepted: 05/15/2021] [Indexed: 12/19/2022]
Abstract
In central serous chorioretinopathy (CSC), the macula is detached because of fluid leakage at the level of the retinal pigment epithelium. The fluid appears to originate from choroidal vascular hyperpermeability, but the etiology for the fluid is controversial. The choroidal vascular findings as elucidated by recent optical coherence tomography (OCT) and wide-field indocyanine green (ICG) angiographic evaluation show eyes with CSC have many of the same venous patterns that are found in eyes following occlusion of the vortex veins or carotid cavernous sinus fistulas (CCSF). The eyes show delayed choroidal filling, dilated veins, intervortex venous anastomoses, and choroidal vascular hyperpermeability. While patients with occlusion of the vortex veins or CCSF have extraocular abnormalities accounting for the venous outflow problems, eyes with CSC appear to have venous outflow abnormalities as an intrinsic phenomenon. Control of venous outflow from the eye involves a Starling resistor effect, which appears to be abnormal in CSC. Similar choroidal vascular abnormalities have been found in peripapillary pachychoroid syndrome. However, peripapillary pachychoroid syndrome has intervortex venous anastomoses located in the peripapillary region while in CSC these are seen to be located in the macular region. Spaceflight associated neuro-ocular syndrome appears to share many of the pathophysiologic problems of abnormal venous outflow from the choroid along with a host of associated abnormalities. These diseases vary according to their underlying etiologies but are linked by the venous decompensation in the choroid that leads to significant vision loss. Choroidal venous overload provides a unifying concept and theory for an improved understanding of the pathophysiology and classification of a group of diseases to a greater extent than previous proposals.
Collapse
Affiliation(s)
- Richard F Spaide
- Vitreous, Retina, Macula Consultants of New York, New York, NY, USA.
| | | | - Hidetaka Matsumoto
- Department of Ophthalmology, Gunma University Graduate School of Medicine, Japan.
| | | | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Elon H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands.
| | | | | | | | | | | | | | - Jay Chhablani
- University of Pittsburgh, UPMC Eye Center, Pittsburgh, PA, USA.
| | - Peter M Maloca
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
| |
Collapse
|
13
|
Remes A, Basha D, Frey N, Wagner A, Müller O. Gene transfer to the vascular system: Novel translational perspectives for vascular diseases. Biochem Pharmacol 2020; 182:114265. [DOI: 10.1016/j.bcp.2020.114265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 01/04/2023]
|
14
|
Wu NC, Chen ZC, Feng IJ, Ho CH, Chiang CY, Wang JJ, Chang WT. Severe varicose veins and the risk of mortality: a nationwide population-based cohort study. BMJ Open 2020; 10:e034245. [PMID: 32565451 PMCID: PMC7311034 DOI: 10.1136/bmjopen-2019-034245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Varicose veins (VVs) are common and although considered benign may cause morbidity. However, the association between VV severity and cardiovascular and mortality risks remains unknown. The aim of this study was to investigate the factors associated with overall mortality in patients with VV. METHODS A total of 4644 patients with newly diagnosed VV between 1999 and 2013 were identified from Taiwan's National Health Insurance Database. VV severity was classified from grade 1 to 3 according to the presentation of ulcers or inflammation. Moreover, 9497, 2541 and 5722 age-matched, sex-matched and chronic cardiovascular risk factor-matched controls, as assessed based on propensity score, were separately selected for three grading VV groups. Enrolled patients were analysed using conditional Cox proportional hazards regression analysis to estimate risk of mortality and major adverse cardiovascular events (MACEs) in the VV and control groups. RESULTS Most patients with VV were free from systemic disease. However, compared with matched controls, patients with VV showed a 1.37 times increased risk of mortality (95% CI 1.19 to 1.57; p<0.0001). Compared with matched controls, older (age ≧65 years) (adjusted HR: 1.38; 95% CI 1.17 to 1.62; p=0.0001) and male patients with VV (adjusted HR 1.41; 95% CI 1.18 to 1.68; p=0.0001) showed increased risk of mortality. Furthermore, compared with controls, patients with VV showed 2.05 times greater risk of MACE. Compared with matched controls, population at grade 3 increased 1.83 times risk of mortality and 2.04 to 38.42 times risk of heart failure, acute coronary syndrome, ischaemic stroke and venous thromboembolism. CONCLUSIONS This nationwide cohort study demonstrated that patients with VV are at a risk of cardiovascular events and mortality. Our findings suggest that presence of VV warrants close attention in terms of prognosis and treatment.
Collapse
Affiliation(s)
- Nan-Chun Wu
- Department of Cardiovascular Surgery, Chi Mei Medical Center, Tainan, Taiwan
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Rende, Taiwan
| | - Zhih-Cherng Chen
- Department of Cardiology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Rende, Taiwan
| | - I-Jung Feng
- Department of Healthcare Administration and Medical Informatics, Chi Mei Medical Center, Tainan, Taiwan
| | - Chung-Han Ho
- Department of Healthcare Administration and Medical Informatics, Chi Mei Medical Center, Tainan, Taiwan
| | - Chun-Yen Chiang
- Department of Cardiology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Rende, Taiwan
| | - Jhi-Joung Wang
- Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Wei-Ting Chang
- Department of Cardiology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
15
|
Studennikova VV, Severgina LO, Sinyavin GV, Rapoport LM, Korovin IA. [Venous wall weackness pathogenesis in varicose vein disease]. Khirurgiia (Mosk) 2019:69-74. [PMID: 31626242 DOI: 10.17116/hirurgia201910169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Varicose veins of lower extremities, varicocele and varicose dilation of pelvic veins including ovaricovaricocele are the most common non-inflammatory diseases of venous system. The same mechanisms determined varicose dilatation regardless localization of the vein. First of all, these are wall weakness and valvular insufficiency combined with hereditary collagen fiber defectiveness in case of undifferentiated connective tissue syndrome. Imbalance between different types of collagen in vascular wall (especially types I and III) also results wall weakness. Other important mechanisms are smooth muscle cells dysfunction followed by excessive intracellular synthesis and intramuscular fibrosis and imbalance of protease system due to overproduction of metalloproteinases. We consider that different forms of varicose veins (varicose veins of lower extremities, varicocele, ovaricovaricocele) may be unified within one pathology (varicose vein disease).
Collapse
Affiliation(s)
- V V Studennikova
- Sechenov First Moscow State Medical University, Ministry of Health of the Russia, Moscow, Russia
| | - L O Severgina
- Sechenov First Moscow State Medical University, Ministry of Health of the Russia, Moscow, Russia
| | - G V Sinyavin
- Sechenov First Moscow State Medical University, Ministry of Health of the Russia, Moscow, Russia
| | - L M Rapoport
- Sechenov First Moscow State Medical University, Ministry of Health of the Russia, Moscow, Russia
| | - I A Korovin
- Sechenov First Moscow State Medical University, Ministry of Health of the Russia, Moscow, Russia
| |
Collapse
|
16
|
Biranvand AS, Khosravi M, Esfandiari G, Poursaleh A, Hosseini-Fard SR, Amirfarhangi A, Najafi M. Associations between miR-661, miR-1202, lncRNA-HOTAIR, lncRNA-GAS5 and MMP9 in differentiated M2-macrophages of patients with varicose veins. INT ANGIOL 2019; 37:451-456. [PMID: 30558403 DOI: 10.23736/s0392-9590.18.04022-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The venous hypertension is suggested as the main cause of varicose disease. Some mediators and growth factors are known as the responsible of cellular events for the progression of venous perturbations. The aim of this study was to investigate non-coding (nc) RNA and MMP9 expression levels in macrophages differentiated from monocytes of patients with varicose veins. METHODS The monocytes were isolated from the whole blood samples by RosetteSep kit and were differentiated to macrophages M2 using M-CSF factor. The based on ncRNA-gene network, lncRNA-GAS5, lncRNA-HOTAIR, miRNA-661, miRNA-1202, and MMP9 were selected. The gene expression levels were measured by RT-qPCR technique. RESULTS Data showed that the MMP9 gene expression increased (P=0.003) while the GAS5, miRNA-661, and miRNA-1202 expression levels reduced significantly in the differentiated macrophages of patients (P=0.035, P=0.009, and P=0.015, respectively). Furthermore, the MMP9 gene expression levels were conversely related to the GAS5, HOTAIR, miRNA-661 and miRNA-1202 expression levels. CONCLUSIONS The results suggested that the lncRNA-GAS5, miRNA-661, miRNA-1202 and MMP9 are involved in varicose disease.
Collapse
Affiliation(s)
- Azin S Biranvand
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Khosravi
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran
| | - Golnaz Esfandiari
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran
| | - Adeleh Poursaleh
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran
| | - Sayed R Hosseini-Fard
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Najafi
- Cellular and Molecular Research Center, Biochemistry Department, Iran University of Medical Sciences, Tehran, Iran - .,Firoozabadi Hospital, Tehran, Iran
| |
Collapse
|
17
|
Affiliation(s)
- Jan Stana
- Specialist in Vascular Surgery; Schön Klinik Vogtareuth, Klinik für operative und interventionelle Gefäßchirurgie, Krankenhausstraße 20, DE-83569 Vogtareuth, Germany
| | - Uroš Maver
- Head of Institute of Biomedical Sciences, Assistant Professor in Pharmacology and Toxicology; University of Maribor, Faculty of Medicine, Institute of Biomedical Sciences, Taborska ulica 8, SI-2000 Maribor, Slovenia. University of Maribor, Faculty of Medicine, Department of Pharmacology, Taborska ulica 8, SI-2000 Maribor, Slovenia
| | - Uroš Potočnik
- Head of Center for Human Molecular Genetics and Pharmacogenomics, Professor of Biochemistry and Genetics; University of Maribor, Faculty of Medicine, Center for human molecular genetics and pharmacogenomics, Taborska ulica 8, SI-2000 Maribor, Slovenia. University of Maribor, Faculty for Chemistry and Chemical engineering, Laboratory for Biochemistry, Molecular Biology and Genomics, Smetanova 17, SI-2000 Maribor, Slovenia
| |
Collapse
|
18
|
Yetkin E, Ozturk S. Dilating Vascular Diseases: Pathophysiology and Clinical Aspects. Int J Vasc Med 2018; 2018:9024278. [PMID: 30225143 PMCID: PMC6129317 DOI: 10.1155/2018/9024278] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/29/2018] [Indexed: 01/22/2023] Open
Abstract
Atherosclerotic disease of the vessels is a significant problem affecting mortality and morbidity all over the world. However, dilatation of the vessels either in the arterial system or in the venous territory is another vessel disease. Varicocele, pelvic, and peripheral varicose veins and hemorrhoids are aneurysms of the venous vascular regions and have been defined as dilating venous disease, recently. Coronary artery ectasia, intracranial aneurysm, and abdominal aortic aneurysm are examples of arterial dilating vascular diseases. Mostly, they have been defined as variants of atherosclerosis. Although there are some similarities in terms of pathogenesis, they are distinct from atherosclerotic disease of the vessels. In addition, pathophysiological and histological similarities and clinical coexistence of these diseases have been demonstrated both in the arterial and in the venous system. This situation underlies the thought that dilatation of the vessels in any vascular territory should be considered as a systemic vessel wall disease rather than being a local disease of any vessel. These patients should be evaluated for other dilating vascular diseases in a systematic manner.
Collapse
Affiliation(s)
- Ertan Yetkin
- Private Yenisehir Hospital, Department of Cardiology, Mersin, Turkey
| | - Selcuk Ozturk
- Ankara Education and Research Hospital, Department of Cardiology, Ankara, Turkey
| |
Collapse
|
19
|
Al-Zoubi NA, Yaghan RJ, Mazahreh TS, Hijazi EM, Alqudah A, Owaisy YN, Hamouri S, Al-Shatnawi NJ. Evaluation of Plasma Growth Factors (VEGF, PDGF, EGF, ANG1, and ANG2) in Patients with Varicose Veins Before and After Treatment with Endovenous Laser Ablation. Photomed Laser Surg 2018; 36:169-173. [DOI: 10.1089/pho.2017.4355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Nabil A. Al-Zoubi
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Rami J. Yaghan
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Tagleb S. Mazahreh
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Emad M. Hijazi
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Yasmin N. Owaisy
- Department of Laboratories, King Abdullah University Hospital (KAUH), Irbid, Jordan
| | - Shadi Hamouri
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nawaf J. Al-Shatnawi
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
20
|
Castro-Ferreira R, Cardoso R, Leite-Moreira A, Mansilha A. The Role of Endothelial Dysfunction and Inflammation in Chronic Venous Disease. Ann Vasc Surg 2018; 46:380-393. [DOI: 10.1016/j.avsg.2017.06.131] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/15/2017] [Accepted: 06/21/2017] [Indexed: 12/14/2022]
|
21
|
Serralheiro P, Novais A, Cairrão E, Maia C, Costa Almeida CM, Verde I. Variability of MMP/TIMP and TGF-β1 Receptors throughout the Clinical Progression of Chronic Venous Disease. Int J Mol Sci 2017; 19:ijms19010006. [PMID: 29267209 PMCID: PMC5795958 DOI: 10.3390/ijms19010006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 01/30/2023] Open
Abstract
Chronic venous disease (CVeD) is a prevalent condition with a significant socioeconomic burden, yet the pathophysiology is only just beginning to be understood. Previous studies concerning the dysregulation of matrix metalloproteinases (MMPs) and their inhibitors (tissue inhibitors of metalloproteinases (TIMPs)) within the varicose vein wall are inconsistent and disregard clinical progression. Moreover, it is highly plausible that MMP and TIMP expression/activity is affected by transforming growth factor (TGF)-β1 and its signaling receptors (TGFβRs) expression/activity in the vein wall. A case–control study was undertaken to analyze genetic and immunohistochemical differences between healthy (n = 13) and CVeD (early stages: n = 19; advanced stages: n = 12) great saphenous vein samples. Samples were grouped based on anatomic harvest site and subjected to quantitative polymerase chain reaction for MMP1, MMP2, MMP8, MMP9, MMP12, MMP13, TIMP1, TIMP2, TIMP3, TIMP4, TGFβR1, TGFβR2, and TGFβR3 gene expression analysis, and then to immunohistochemistry for immunolocalization of MMP2, TIMP2, and TGFβR2. Decreased gene expression of MMP12, TIMP2, TIMP3, TIMP4, and TGFβR2 was found in varicose veins when compared to controls. Regarding CVeD clinical progression, two facts arose: results across anatomical regions were uneven; decreased gene expression of MMP9 and TGFβR3 and increased gene expression of MMP2 and TIMP3 were found in advanced clinical stages. Most immunohistochemistry results for tunica intima were coherent with qPCR results. In conclusion, decreased expression of TGFβRs might suggest a reduction in TGF-β1 participation in the MMP/TIMP imbalance throughout CVeD progression. Further studies about molecular events in the varicose vein wall are required and should take into consideration the venous anatomical region and CVeD clinical progression.
Collapse
Affiliation(s)
- Pedro Serralheiro
- Norfolk and Norwich University Hospital, Norwich NR47UY, UK.
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| | - António Novais
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| | - Elisa Cairrão
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| | - Cláudio Maia
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| | - Carlos M Costa Almeida
- Department of General Surgery (C), Coimbra University Hospital Centre, 3041-801 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Ignacio Verde
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| |
Collapse
|
22
|
Serralheiro P, Soares A, Costa Almeida CM, Verde I. TGF-β1 in Vascular Wall Pathology: Unraveling Chronic Venous Insufficiency Pathophysiology. Int J Mol Sci 2017; 18:E2534. [PMID: 29186866 PMCID: PMC5751137 DOI: 10.3390/ijms18122534] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022] Open
Abstract
Chronic venous insufficiency and varicose veins occur commonly in affluent countries and are a socioeconomic burden. However, there remains a relative lack of knowledge about venous pathophysiology. Various theories have been suggested, yet the molecular sequence of events is poorly understood. Transforming growth factor-beta one (TGF-β1) is a highly complex polypeptide with multifunctional properties that has an active role during embryonic development, in adult organ physiology and in the pathophysiology of major diseases, including cancer and various autoimmune, fibrotic and cardiovascular diseases. Therefore, an emphasis on understanding its signaling pathways (and possible disruptions) will be an essential requirement for a better comprehension and management of specific diseases. This review aims at shedding more light on venous pathophysiology by describing the TGF-β1 structure, function, activation and signaling, and providing an overview of how this growth factor and disturbances in its signaling pathway may contribute to specific pathological processes concerning the vessel wall which, in turn, may have a role in chronic venous insufficiency.
Collapse
Affiliation(s)
- Pedro Serralheiro
- Norfolk and Norwich University Hospital, Colney Ln, Norwich NR47UY, UK.
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| | - Andreia Soares
- Norfolk and Norwich University Hospital, Colney Ln, Norwich NR47UY, UK.
| | - Carlos M Costa Almeida
- Department of General Surgery (C), Coimbra University Hospital Centre, Portugal; Faculty of Medicine, University of Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal.
| | - Ignacio Verde
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| |
Collapse
|
23
|
Alqudah M, Qandeel H, Al-Zoubi N, Alqudah A, Bani-Ahmad M, Alzoubi A. Changes of serum growth factors profiles in patients with venous thromboembolism. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 77:595-600. [DOI: 10.1080/00365513.2017.1379607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mohammad Alqudah
- Department of Pathology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Haitham Qandeel
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nabil Al-Zoubi
- Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Mohammad Bani-Ahmad
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Abdallah Alzoubi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
24
|
Jacobs BN, Andraska EA, Obi AT, Wakefield TW. Pathophysiology of varicose veins. J Vasc Surg Venous Lymphat Disord 2017; 5:460-467. [DOI: 10.1016/j.jvsv.2016.12.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 12/27/2016] [Indexed: 01/09/2023]
|
25
|
Kun L, Ying L, Lei W, Jianhua Z, Yongbo X, Tao W, Jinyuan T, Haibo C. Dysregulated apoptosis of the venous wall in chronic venous disease and portal hypertension. Phlebology 2016; 31:729-736. [PMID: 26447135 DOI: 10.1177/0268355515610237] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction The etiology of varicose veins remains elusive. We hypothesized that abnormal cell cycle events in the vein wall may contribute to changes in the structural integrity, thus predisposing to the development of varicosities. The present study was designed to determine whether or not the same molecular apoptotic pathway exists between great saphenous and splenic veins. Methods Thirty-six samples of diseased splenic veins and varicose great saphenous veins were collected. Twenty-five samples of control splenic and great saphenous veins were also collected. The apoptotic cell proteins expression was immunohistochemically stained with antibodies (anti-Bax and anti-Bcl-xl). Apoptosis was evaluated by the terminal deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL) assay and immunofluorescence staining. The morphology of apoptotic cells was observed with an electron microscope. Results The apoptotic ratio in walls (intima and media) of diseased splenic vein and varicose great saphenous vein groups were significantly lower than the corresponding regions in the splenic vein and great saphenous vein groups ( p < 0.01), respectively. A significant difference was not noted in the ratio change of apoptotic cells between the diseased splenic vein and varicose great saphenous vein groups ( p > 0.05). The high positive expression of Bcl-xl proteins was detected in the diseased splenic vein and varicose great saphenous vein groups, respectively. While the high positive expression of Bax proteins was also observed in the splenic vein and great saphenous vein groups, respectively. Electron microscopic observations confirmed that endothelial and smooth muscle cells in diseased splenic vein, varicose great saphenous vein, splenic vein, and great saphenous vein walls exhibited apoptotic morphologic features, such as fuzzy mitochondrial cristae, medullary changes, and margination of the nuclear chromatin. Conclusions Our results showed the same dysregulation of apoptosis via the intrinsic pathway in diseased splenic veins and varicose great saphenous veins. This observational study suggests that apoptotic down-regulation in the veins wall is a cause of diseased splenic veins and varicose great saphenous veins, but does not exclude the possibility that other mechanisms are involved.
Collapse
Affiliation(s)
- Li Kun
- Department of General Surgery, 89th Hospital of PLA, Weifang, China
| | - Li Ying
- Department of General Surgery, 89th Hospital of PLA, Weifang, China
| | - Wang Lei
- Department of Postgraduate, Weifang Medical College, Weifang, China
| | - Zhao Jianhua
- Department of General Surgery, 89th Hospital of PLA, Weifang, China
| | - Xu Yongbo
- Department of General Surgery, 89th Hospital of PLA, Weifang, China
| | - Wang Tao
- Department of Pathology, 89th Hospital of PLA, Weifang, China
| | - Tang Jinyuan
- Department of General Surgery, 89th Hospital of PLA, Weifang, China
| | - Chu Haibo
- Department of General Surgery, 89th Hospital of PLA, Weifang, China
| |
Collapse
|
26
|
Naoum JJ, Hunter GC. Pathogenesis of Varicose Veins and Implications for Clinical Management. Vascular 2016; 15:242-9. [DOI: 10.2310/6670.2007.00069] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Varicose veins (VVs) classically result from venous hypertension owing to incompetence of the major communications between the superficial and deep veins of the lower extremity. In a significant number of patients, there is no demonstrable truncal saphenous reflux and varicosities are the result of isolated perforating and nonsaphenous vein incompetence. The clinical and histologic features of VVs are the result of disruption of the normal architectural structure of the venous wall as a consequence of remodeling of the extracellular matrix (ECM) in response to increased venous distention and altered hemodynamic shear stress. Although a number of genes, growth factors, proteases, and their inhibitors known to modulate the ECM have been implicated in the pathogenesis of VVs, their etiology remains unknown. The complex variations in venous anatomy in patients with VVs require detailed vein mapping to determine the source and drainage locations of reflux if the rates of residual and recurrent varicosities are to be reduced. The distinct pathogenic mechanisms involved in the development of VVs have important implications for the management of VVs that include a wide spectrum of treatment modalities ranging from reassurance, alternative medicines, conservative management or compression therapy, and surgical or endovascular therapy.
Collapse
Affiliation(s)
- Joseph J. Naoum
- *The Methodist Hospital, Methodist DeBakey Heart Center, Houston, TX; †Carl T. Hayden VA Medical Center, Phoenix, AZ
| | - Glenn C. Hunter
- *The Methodist Hospital, Methodist DeBakey Heart Center, Houston, TX; †Carl T. Hayden VA Medical Center, Phoenix, AZ
| |
Collapse
|
27
|
Serralheiro P, Cairrão E, Maia CJ, João M, Almeida CMC, Verde I. Effect of TGF-beta1 on MMP/TIMP and TGF-beta1 receptors in great saphenous veins and its significance on chronic venous insufficiency. Phlebology 2016; 32:334-341. [DOI: 10.1177/0268355516655067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objectives Transforming growth factor-beta1 (TGF-β1) may participate in local chronic inflammatory processes in varicose veins and in venous wall structure modifications through regulation of matrix metalloproteinases (MMP) and their inhibitors (tissue inhibitor of metalloproteinase (TIMP)). The aim of this study was to analyze the effect of TGF-β1 in the vein wall, namely on the gene expression of selected MMP, TIMP and TGF-β1 receptors. Methods Healthy vein samples were harvested from eight subjects who underwent coronary bypass graft surgery with great saphenous vein. Each vein sample was divided into two segments, which were cultivated separately in vitro (one of the segments had TGF-β1 added) and then submitted to gene expression analysis. Results In the TGF-β1 supplemented group, there was a general increase in the mean gene expression. Specifically, expression of MMP9, MMP12, TIMP1 and TIMP2 were statistically significant. Conclusion The results of this study demonstrate that the gene expression of MMP9, MMP12, TIMP1 and TIMP2 was influenced by the addition of TGF-β1. These results may be translated to chronic venous insufficiency framework and suggest involvement of TGF-β1 in the vein wall pathology.
Collapse
Affiliation(s)
- Pedro Serralheiro
- Department of General Surgery, Norfolk and Norwich University Hospital, UK
- Faculdade de Ciências da Saúde, University of Beira Interior, Portugal
| | - Elisa Cairrão
- Faculdade de Ciências da Saúde, University of Beira Interior, Portugal
| | - Cláudio J Maia
- Faculdade de Ciências da Saúde, University of Beira Interior, Portugal
| | - Marina João
- Faculdade de Ciências da Saúde, University of Beira Interior, Portugal
| | - Carlos M Costa Almeida
- Department of General Surgery, Coimbra University Hospital Centre, Portugal
- Faculdade de Medicina, University of Coimbra, Portugal
| | - Ignacio Verde
- Faculdade de Ciências da Saúde, University of Beira Interior, Portugal
| |
Collapse
|
28
|
Yetkin E, Ileri M. Dilating venous disease: Pathophysiology and a systematic aspect to different vascular territories. Med Hypotheses 2016; 91:73-76. [PMID: 27142148 DOI: 10.1016/j.mehy.2016.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 12/13/2022]
Abstract
Venous disease is a common but overlooked clinical problem and is an important mortality and morbidity factor depending on the effected vascular territory. Different contributing factors play role on the clinical manifestation of the disease. Peripheral varices of lower extremities, hemorrhoids, varicoceles, pelvic varicose veins are the vasculopathy of veins running toward heart but against gravity. We hypothesize that all these clinical entities share common pathophysiologic steps in terms of vascular wall remodeling and vessel wall damage. A systematic approaches to both arterial and venous dilating disease in further studies and research would increase our understanding on the pathophysiology of dilating vascular disease and would provoke to find out new treatment modalities. Varicose remodeling of veins occurs by a complex interplay of various factors including both physical forces and extracellular matrix remodeling mechanisms. This article focuses on the systematic aspects of dilating venous disease with a focus on pathophysiology under the term of "Dilating Venous Disease".
Collapse
Affiliation(s)
- Ertan Yetkin
- Yenisehir Hospital, Division of Cardiology, Mersin, Turkey.
| | - Mehmet Ileri
- Numune Training and Research Hospital, Department of Cardiology, Ankara, Turkey
| |
Collapse
|
29
|
Decano JL, Pasion KA, Black N, Giordano NJ, Herrera VL, Ruiz-Opazo N. Sex-specific genetic determinants for arterial stiffness in Dahl salt-sensitive hypertensive rats. BMC Genet 2016; 17:19. [PMID: 26754450 PMCID: PMC4709875 DOI: 10.1186/s12863-015-0324-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/22/2015] [Indexed: 02/08/2023] Open
Abstract
Background Arterial stiffness is an independent predictor of cardiovascular outcomes in hypertensive patients including myocardial infarction, fatal stroke, cerebral micro-bleeds which predicts cerebral hemorrhage in hypertensive patients, as well as progression to hypertension in non-hypertensive subjects. The association between arterial stiffness and various cardiovascular outcomes (coronary heart disease, stroke) remains after adjusting for age, sex, blood pressure, body mass index and other known predictors of cardiovascular disease, suggesting that arterial stiffness, measured via carotid-femoral pulse wave velocity, has a better predictive value than each of these factors. Recent evidence shows that arterial stiffening precedes the onset of high blood pressure; however their molecular genetic relationship (s) and sex-specific determinants remain uncertain. We investigated whether distinct or shared genetic determinants might underlie susceptibility to arterial stiffening in male and female Dahl salt-sensitive rats. Thus, we performed a genome-wide scan for quantitative trait loci (QTLs) affecting arterial stiffness in six-week old F2 (Dahl S x R)-intercross male and female rats characterized for abdominal aortic pulse wave velocity and aortic strain by high-resolution ultrasonography. Results We detected five highly significant QTLs affecting aortic stiffness: two interacting QTLs (AS-m1 on chromosome 4 and AS-m2 on chromosome16, LOD 8.8) in males and two distinct interacting QTLs (AS-f1 on chromosome 9 and AS-f2 on chromosome11, LOD 8.9) in females affecting pulse wave velocity. One QTL (AS-1 on chromosome 3, LOD 4.3) was found to influence aortic strain in a sex-independent manner. None of these arterial stiffness QTLs co-localized with previously reported blood pressure QTLs detected in equivalent genetic intercrosses. Conclusions These data reveal sex-specific genetic determinants for aortic pulse wave velocity and suggest distinct polygenic susceptibility for arterial stiffness and salt-sensitive hypertension in Dahl rats based upon reported blood pressure QTLs in equivalent (Dahl S x R)-intercrosses.
Collapse
Affiliation(s)
- Julius L Decano
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany Street, W-609, Boston, MA, 02118, USA.
| | - Khristine A Pasion
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany Street, W-609, Boston, MA, 02118, USA.
| | - Nicole Black
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany Street, W-609, Boston, MA, 02118, USA.
| | - Nicholas J Giordano
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany Street, W-609, Boston, MA, 02118, USA.
| | - Victoria L Herrera
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany Street, W-609, Boston, MA, 02118, USA.
| | - Nelson Ruiz-Opazo
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany Street, W-609, Boston, MA, 02118, USA.
| |
Collapse
|
30
|
Ozturk S, Yalvac HD, Sivri N, Ozturk HM, Kılıc Y, Bulut E, Celik A, Barlas Y, Tengiz I, Yetkin E. Anxiety and depression scores in patients with coronary artery disease and coronary artery ectasia. Int J Cardiol 2015; 186:299-301. [PMID: 25828141 DOI: 10.1016/j.ijcard.2015.03.305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/30/2022]
Affiliation(s)
- S Ozturk
- Abant Izzet Baysal University Izzet Baysal Research and Training Hospital, Department of Cardiology, Bolu, Turkey
| | - H D Yalvac
- DR Abdurrahman Yurtaslan Oncology Research and Training Hospital, Department of Psychiatry, Ankara, Turkey
| | - N Sivri
- Trakya University Faculty of Medicine, Department of Cardiology, Edirne Turkey
| | - H M Ozturk
- Abant Izzet Baysal University Izzet Baysal Research and Training Hospital, Department of Psychiatry, Bolu, Turkey
| | - Y Kılıc
- Abant Izzet Baysal University Izzet Baysal Research and Training Hospital, Department of Cardiology, Bolu, Turkey
| | - E Bulut
- Abant Izzet Baysal University Izzet Baysal Research and Training Hospital, Department of Cardiology, Bolu, Turkey
| | - A Celik
- Abant Izzet Baysal University Izzet Baysal Research and Training Hospital, Department of Cardiology, Bolu, Turkey
| | - Y Barlas
- Abant Izzet Baysal University Izzet Baysal Research and Training Hospital, Department of Cardiology, Bolu, Turkey
| | - I Tengiz
- Izmir University Medical Park Hospital, Department of Cardiology, Izmir, Turkey
| | - E Yetkin
- Middle East Hospital, Department of Cardiology, Mersin, Turkey.
| |
Collapse
|
31
|
Pocock ES, Alsaigh T, Mazor R, Schmid-Schönbein GW. Cellular and molecular basis of Venous insufficiency. Vasc Cell 2014; 6:24. [PMID: 25520775 PMCID: PMC4268799 DOI: 10.1186/s13221-014-0024-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/20/2014] [Indexed: 01/30/2023] Open
Abstract
Chronic venous disease (CVD) has a range of clinical presentations, including tortuous, distended veins in lower extremities, increasing skin pigmentation, and in severe cases ulceration of the affected skin. Venous insufficiency, a precursor to CVD characterized by improper return of blood from the lower extremities to the heart, must be studied in its earliest stages at a time when preventative measures could be applied in man. This underscores the need for basic research into biomarkers and genetic predisposing factors affecting the progression of venous disease. Investigation over the past decade has yielded insight into these specific genetic, cellular and molecular mechanisms underlying the development of venous disease. Among the many advances include the elucidation of an increasing role for matrix metalloproteinases as important mediators of the degenerative process involved with venous insufficiency. This may be preceded by an inflammatory process which further contributes to venular degeneration and endothelial dysfunction seen in advanced presentation of disease. Furthermore, genomic analyses have shed light upon temporal expression patterns of matrix remodeling proteins in diseased tissue samples. In this review we examine some of the current findings surrounding cellular, molecular and genetic advances in delineating the etiology of chronic venous disease.
Collapse
Affiliation(s)
- Elizabeth S Pocock
- Department of Bioengineering, The Institute for Engineering in Medicine, University of California San Diego, 92093-0412 La Jolla, California
| | - Tom Alsaigh
- Department of Bioengineering, The Institute for Engineering in Medicine, University of California San Diego, 92093-0412 La Jolla, California
| | - Rafi Mazor
- Department of Bioengineering, The Institute for Engineering in Medicine, University of California San Diego, 92093-0412 La Jolla, California
| | - Geert W Schmid-Schönbein
- Department of Bioengineering, The Institute for Engineering in Medicine, University of California San Diego, 92093-0412 La Jolla, California
| |
Collapse
|
32
|
Ketchum LD. The Rationale for Treating the Nodule in Dupuytren's Disease. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2014; 2:e278. [PMID: 25587512 PMCID: PMC4292260 DOI: 10.1097/gox.0000000000000249] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 11/06/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Dupuytren's disease encompasses a spectrum of fibroblastic disorders from the presence of 1-2 nodules in the palm of a hand with no joint contracture to grotesque, crippling and disabling deformities of hands. Over the last 50 years, many enlightening studies have been forthcoming, which, through techniques of histopathology, biochemistry, tissue culture, and electron microscopy, have shed pearls of light on various aspects of the disease process. METHODS A comprehensive review of the literature on Dupuytren's disease was undertaken, stringing together results from numerous studies to establish a table of events and their location in the development of the disease. RESULTS What will be seen is a credible scenario of events which will enable the clinician to be more proactive in the earlier treatment of the disease and more aware of factors that increase or decrease recurrence rates postoperatively. CONCLUSIONS The most effective management of Dupuytren's disease is early recognition and treatment of the nodule, before the development of a joint contracture, particularly of a proximal interphalangeal joint. As there is evidence of a significant inflammatory role in the development of the nodule, the process of fibroplasia can be minimized by altering the macrophage > fibroblast > collagen cascade by the intralesional injection of a potent anti-inflammatory agent such as triamcinolone, which also blocks tissue inhibitors of collagenase, thus enhancing the action of native collagenase, and reduces the size and firmness of nodules and, at least temporarily, arrests their progression.
Collapse
Affiliation(s)
- Lynn D Ketchum
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, Kans
| |
Collapse
|
33
|
Abstract
Recent sequencing of the human genome has opened up new areas of investigation for genetic aberrations responsible for the pathogenesis of many human diseases. To date, there have been no studies that have investigated the entire human genome for the genetic underpinnings of chronic venous insufficiency (CVI). Utilizing Gene Chip Arrays we analyzed the relative expression levels of more than 47,000 transcripts and variants and approximately 38,500 well-characterized genes from each of 20 patients (N (CVI)=10; N (Control Group)=10). Relative gene expression profiles significantly differed between patients with CVI and patients unaffected by CVI. Regulatory genes of mediators of the inflammatory reaction and collagen production were up-regulated and down-regulated, respectively in CVI patients. DNA microarray analysis also showed that relative gene expression of multiple genes which function remains to be elucidated was significantly different in CVI patients. Fundamental advancements in our knowledge of the human genome and understanding of the genetic basis of CVI represents an opportunity to develop new diagnostic, prognostic, preventive and therapeutic modalities in the management of CVI.
Collapse
Affiliation(s)
- Jovan N Markovic
- Department of Vascular Surgery, Duke University School of Medicine, Durham, NC 27710, USA.
| | | |
Collapse
|
34
|
Alexander JS, Prouty L, Tsunoda I, Ganta CV, Minagar A. Venous endothelial injury in central nervous system diseases. BMC Med 2013; 11:219. [PMID: 24228622 PMCID: PMC3851779 DOI: 10.1186/1741-7015-11-219] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 09/09/2013] [Indexed: 02/08/2023] Open
Abstract
The role of the venous system in the pathogenesis of inflammatory neurological/neurodegenerative diseases remains largely unknown and underinvestigated. Aside from cerebral venous infarcts, thromboembolic events, and cerebrovascular bleeding, several inflammatory central nervous system (CNS) diseases, such as multiple sclerosis (MS), acute disseminated encephalomyelitis (ADEM), and optic neuritis, appear to be associated with venous vascular dysfunction, and the neuropathologic hallmark of these diseases is a perivenous, rather than arterial, lesion. Such findings raise fundamental questions about the nature of these diseases, such as the reasons why their pathognomonic lesions do not develop around the arteries and what exactly are the roles of cerebral venous inflammation in their pathogenesis. Apart from this inflammatory-based view, a new hypothesis with more focus on the hemodynamic features of the cerebral and extracerebral venous system suggests that MS pathophysiology might be associated with the venous system that drains the CNS. Such a hypothesis, if proven correct, opens new therapeutic windows in MS and other neuroinflammatory diseases. Here, we present a comprehensive review of the pathophysiology of MS, ADEM, pseudotumor cerebri, and optic neuritis, with an emphasis on the roles of venous vascular system programming and dysfunction in their pathogenesis. We consider the fundamental differences between arterial and venous endothelium, their dissimilar responses to inflammation, and the potential theoretical contributions of venous insufficiency in the pathogenesis of neurovascular diseases.
Collapse
Affiliation(s)
- Jonathan S Alexander
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Leonard Prouty
- Department of Pathology, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Ikuo Tsunoda
- Department of Microbiology and Immunology, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Chaitanya Vijay Ganta
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Alireza Minagar
- Department of Neurology, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| |
Collapse
|
35
|
CD68 expression in aortocoronary saphenous vein bypass grafts. Histochem Cell Biol 2012; 140:183-8. [PMID: 23275124 DOI: 10.1007/s00418-012-1069-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2012] [Indexed: 10/27/2022]
Abstract
Atherosclerosis commonly affects the arteries harvested from patients 70 years of age or older. Saphenous vein grafts appear to maintain a higher patency rate after coronary artery bypass grafting in these subjects. The infiltration of macrophages is an early step in saphenous vein graft atherosclerosis; however, little is known regarding the underlying mechanisms of infiltration. The objective of the present report is to evaluate the presence of CD68-positive cells in the saphenous vein wall and correlate initial CD68-positive infiltration to specific clinical and biochemical parameters and the graft patency rate as estimated in patients undergoing coronary artery bypass grafting. A total of 309 patients were allocated into two groups: A1 patients, who were between 50 and 70 years of age, and A2 patients, who were 70 years or older at the time of vein harvesting. CD68 expression was evaluated by immunohistochemistry. There were no significant differences between A1 and A2 patients regarding macrophage expression within any of the analyzed vascular regions. Saphenous vein macrophages were never present in the tunica intima unless they were also expressed in the media or the adventitia. The patients with CD68-positive cells in the tunica intima had a significantly higher number of bypass stenoses when compared with the subjects who did not have CD68-positive cells in this layer. These findings suggest that the CD68-positive cells (those that have not yet developed into foam cells) present in the intima of saphenous vein grafts might serve as a very early marker of graft occlusion.
Collapse
|
36
|
Anwar M, Shalhoub J, Vorkas P, Lim C, Want E, Nicholson J, Holmes E, Davies A. In-vitro Identification of Distinctive Metabolic Signatures of Intact Varicose Vein Tissue via Magic Angle Spinning Nuclear Magnetic Resonance Spectroscopy. Eur J Vasc Endovasc Surg 2012; 44:442-50. [DOI: 10.1016/j.ejvs.2012.05.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 05/19/2012] [Indexed: 01/13/2023]
|
37
|
Varicose veins: role of mechanotransduction of venous hypertension. Int J Vasc Med 2012; 2012:538627. [PMID: 22489273 PMCID: PMC3303599 DOI: 10.1155/2012/538627] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 11/13/2011] [Indexed: 11/17/2022] Open
Abstract
Varicose veins affect approximately one-third of the adult population and result in significant psychological, physical, and financial burden. Nevertheless, the molecular pathogenesis of varicose vein formation remains unidentified. Venous hypertension exerted on veins of the lower extremity is considered the principal factor in varicose vein formation. The role of mechanotransduction of the high venous pressure in the pathogenesis of varicose vein formation has not been adequately investigated despite a good progress in understanding the mechanomolecular mechanisms involved in transduction of high blood pressure in the arterial wall. Understanding the nature of the mechanical forces, the mechanosensors and mechanotransducers in the vein wall, and the downstream signaling pathways will provide new molecular targets for the prevention and treatment of varicose veins. This paper summarized the current understanding of mechano-molecular pathways involved in transduction of hemodynamic forces induced by blood pressure and tries to relate this information to setting of venous hypertension in varicose veins.
Collapse
|
38
|
Pathogenesis of Varicose Veins. J Vasc Interv Radiol 2012; 23:33-9; quiz 40. [DOI: 10.1016/j.jvir.2011.09.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 08/31/2011] [Accepted: 09/09/2011] [Indexed: 01/24/2023] Open
|
39
|
Influence of thrombophlebitis on TGF-β1 and its signaling pathway in the vein wall. Folia Histochem Cytobiol 2011; 48:542-8. [PMID: 21478096 DOI: 10.2478/v10042-010-0041-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Extensive extracellular matrix remodeling of the vein wall is involved in varicose veins pathogenesis. This process is controlled by numerous factors, including peptide growth factors. The aim of the study was to evaluate influence of thrombophlebitis on TGF-β1 and its signaling pathway in the vein wall. TGF-β1 mRNAlevels, growth factor content and its expression were evaluated by RT-PCR, ELISA, and western blot methods, respectively, in the walls of normal veins, varicose veins and varicose veins complicated by thrombophlebitis. Western blot analysis was used to assess TGF-β receptor type II (TGF-β RII) and p-Smad2/3 protein expression in the investigated material. Unchanged mRNA levels of TGF-β1, decreased TGF-β1 content, as well as decreased expression of latent and active forms of TGF-β1 were found in varicose veins. Increased expression of TGF-β RII and p-Smad2/3 were found in varicose veins. Thrombophlebitis led to increased protein expression of the TGF-β1 active form and p-Smad2/3 in the vein wall compared to varicose veins. TGF-β1 may play a role in the disease pathogenesis because of increased expression and activation of its receptor in the wall of varicose veins. Thrombophlebitis accelerates activation of TGF-β1 and activity of its receptor in the varicose vein wall.
Collapse
|
40
|
Varicose veins show enhanced chemokine expression. Eur J Vasc Endovasc Surg 2009; 38:635-41. [PMID: 19729323 DOI: 10.1016/j.ejvs.2009.07.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 07/09/2009] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Leucocyte infiltration in the wall of varicose veins has been reported previously. This study was designed to investigate the expression of pro-inflammatory cytokines and chemokines in control and in patients with varicose veins and to test the effect of treating varicose vein patients with acetylsalicylic acid (ASA) on cytokine expression prior to removal of varices. MATERIAL AND METHODS Sections of vein were removed during operation from both patient groups, and ribonuclease protection assays (RPAs) were performed to assess the expression of chemokines. Group I included non-varicose saphenous veins from healthy patients undergoing amputation for trauma. Varicose veins were obtained from patients with primary varicose undergoing surgical treatment who received no drug (group II) or treatment with 300 mg day(-1) of ASA for 15 days before surgery (group III). RESULTS Non-varicose veins constitutively expressed low levels of monocyte-chemoattractant protein (MCP-1) and interleukin (IL)-8 mRNA. Varicose veins had a distinct chemokine expression pattern, since significant up-regulation of MCP-1 and IL-8 and a marked expression of IP-10, RANTES, MIP-1alpha and MIP-1beta mRNA were detected. Removal of the endothelium did not alter this pattern. Varicose veins obtained from patients treated with ASA showed a consistent decrease in chemokine expression, although it did not reach statistical significance. CONCLUSIONS Varicose veins showed increased expression of several chemokines compared to control veins. A non-significant reduction of activation was observed following treatment with ASA for 15 days.
Collapse
|
41
|
Yetkin E, Waltenberger J. Novel insights into an old controversy: is coronary artery ectasia a variant of coronary atherosclerosis? Clin Res Cardiol 2007; 96:331-9. [PMID: 17453130 PMCID: PMC2775118 DOI: 10.1007/s00392-007-0521-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 02/12/2007] [Indexed: 01/26/2023]
Abstract
Coronary artery ectasia (CAE) is defined as a localized or diffuse non-obstructive lesion of the epicardial coronary arteries with a luminal dilation exceeding 1.5-fold the diameter of the normal adjacent arterial segment. The incidence of CAE has been reported to range between 2% and 4%, which might be an overestimation of the true frequency. The coincidence of CAE with other systemic vascular dilatations has suggested that the mechanism underlying CAE is not only localized to coronary arteries, but also to other vascular compartments such as aorta or peripheral veins. Although the pathophysiology of CAE remains largely unknown, it was supposed to represent a variant of coronary atherosclerosis. This review focuses on this controversy of whether CAE and coronary artery disease (CAD) are two manifestations of the same underlying process. There are clear differences between CAD and CAE with respect to cardiovascular risk factors such as diabetes mellitus, and pathogenic steps in disease progress such as inflammation or extracellular matrix remodeling. As this review will underscore, the current knowledge of the field is insufficient to finally clarify the causative interrelation between CAE and CAD. The clinical course and treatment of CAE mainly depends on its coexistence with CAD. When coexisting with CAD, the prognosis and treatment of CAE are the same as for CAD alone. In isolated CAE, prognosis is better and anti-platelet drugs are the mainstay of treatment. Surgical treatment can be considered in selected patients. For clarifying the mechanism underlying CAE, additional clinical, histopathological and pathophysiological investigations are required. In fact, every patient with CAE should be evaluated systematically for pathological changes in other vascular territories, both in the arterial system as well as in the venous system, which might occur in the disease process.
Collapse
Affiliation(s)
- Ertan Yetkin
- Department of Cardiology, University Hospital of Maastricht
and Cardiovascular Research Institute
of Maastricht (CARIM)
University of Maastricht, P. Debyelaan 25, 5800, 6202 AZ Maastricht, The Netherlands
| | - Johannes Waltenberger
- Department of Cardiology, University Hospital of Maastricht
and Cardiovascular Research Institute
of Maastricht (CARIM)
University of Maastricht, P. Debyelaan 25, 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
42
|
Sie MPS, Mattace-Raso FUS, Uitterlinden AG, Arp PP, Hofman A, Hoeks APG, Reneman RS, Asmar R, van Duijn CM, Witteman JCM. TGF-beta1 polymorphisms and arterial stiffness; the Rotterdam Study. J Hum Hypertens 2007; 21:431-7. [PMID: 17361195 DOI: 10.1038/sj.jhh.1002175] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Arterial stiffness is a risk factor for cardiovascular disease. Transforming growth factor beta1 is a pleiotropic cytokine, with many functions, including influence on the vascular wall (e.g., on angiogenesis, endothelial cells and the extracellular matrix). We investigated five functional polymorphisms in the transforming growth factor beta1 gene (-800 G/A, -509 C/T, codon 10 Leu/Pro, codon 25 Arg/Pro and codon 263 Thr/Ile) in relation to arterial stiffness in a population-based study. A total of 3863 participants of the Rotterdam Study, a prospective population-based study, were included in the current study. The relations of the genotypes and haplotypes with arterial stiffness (pulse wave velocity (PWV), distensibility coefficient (DC) and pulse pressure (PP)) were studied using analyses of variance and linear regression. The analyses were adjusted for age, sex, mean arterial pressure, heart rate, conventional cardiovascular risk factors and measures of atherosclerosis. There were no associations between PWV and -800 G/A (P=0.56), -509 C/T (P=0.29), codon 10 (P=0.98) and, codon 25 (P=0.28). These polymorphisms were not associated with the DC or with PP. The haplotype-based analyses yielded similar results. The results of this study show that the TGF-beta1 -800 G/A, -509 C/T, codon 10 Leu/Pro and codon 25 Arg/Pro polymorphisms are not associated with arterial stiffness.
Collapse
Affiliation(s)
- M P S Sie
- Department of Epidemiology and Biostatistics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pascual G, Mendieta C, García-Honduvilla N, Corrales C, Bellón JM, Buján J. TGF-beta1 upregulation in the aging varicose vein. J Vasc Res 2007; 44:192-201. [PMID: 17337905 DOI: 10.1159/000100375] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Accepted: 12/26/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although the etiology of venous insufficiency is not well understood, immune response and aging are beginning to emerge as contributing factors. Factors involved in tissue remodeling such as TGF-beta(1) also seem to play an important role in extracellular matrix production. The aim of this study was to explore the relationship between chronic venous insufficiency and TGF-beta(1) examining the latent/mature form of TGF-beta(1) and the presence of mast cells. Effects of age were also evaluated. METHODS Saphenous veins were obtained from patients subjected to aortocoronary bypass (controls) and undergoing varicose vein surgery. These were immunolabeled using anti-LAP TGF-beta(1)/anti-TGF-beta(1) antibodies and subjected to Western blot. Mast cell population was identified by metachromatic staining. RESULTS Latent TGF-beta(1) was significantly reduced in varicose veins from older subjects. In contrast, smooth muscle cells obtained from the varicosities showed intense levels. Mature TGF-beta(1) significantly differed between healthy and varicose veins. No mature TGF-beta(1) was detected in the cell cultures. Mast cell number and degranulation were increased with aging and varicose disease, colocalizing with the mature form of TGF-beta(1). CONCLUSION Aging and varicose pathology induce dysregulation of TGF-beta(1) that could play an important role in the fibrous process, representing the final stages of venous insufficiency.
Collapse
Affiliation(s)
- Gemma Pascual
- Department of Medical Specialities, Faculty of Medicine, University of Alcala, Alcalá de Henares, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
This article reviews the basic science elements involved in the pathogenesis of Dupuytren's disease. Dupuytren's contracture is effected by a multitude of complex processes at the cellular level and the exact relative contribution of each is unknown. Various investigators have proposed different theories and documented individual findings regarding the pathophysiology of Dupuytren's contracture. The current report attempts to summarize many of these findings together in a schema of pathogenesis.
Collapse
|
45
|
Heffernan D, Dudley B, McNeil PL, Howdieshell TR. Local Arginine Supplementation Results in Sustained Wound Nitric Oxide Production and Reductions in Vascular Endothelial Growth Factor Expression and Granulation Tissue Formation. J Surg Res 2006; 133:46-54. [PMID: 16631200 DOI: 10.1016/j.jss.2006.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Revised: 03/10/2006] [Accepted: 03/10/2006] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The goal of this work was to test the functional role of L-arginine in promotion of nitric oxide (NO) production and the vigorous granulation tissue formation characteristic of this wound model. BACKGROUND Therapeutic use of supplemental arginine has been proposed as a safe and efficacious method to produce NO from nitric oxide synthase (NOS) and to produce proline and polyamines from arginase to improve wound healing. Although NO appears to be necessary to promote wound healing, the preferential metabolism of arginine to NO via NOS 2 may be detrimental if maintained beyond the initial days of healing. METHODS A ventral hernia, surgically created in the abdominal wall of 12 swine, was repaired with silicone sheeting and skin closure. Osmotic infusion pumps, inserted in remote subcutaneous pockets, continuously delivered saline (n = 6) or L-arginine (n = 6) into the wound environment. Granulation tissue thickness was determined by ultrasonography. Fluid was aspirated serially from the wound compartment for measurements of nitrite/nitrate (NOx), vascular endothelial growth factor (VEGF), transforming growth factor-beta1 (TGF-beta1), and amino acid concentrations. On day 14, the animals were sacrificed and the abdominal wall was harvested for immunohistochemical and molecular analysis. RESULTS In animals receiving saline, a nearly linear four-fold increase in granulation tissue thickness was measured during the 14-day interval. In contrast, quantitative ultrasound analysis detected significant reductions in L-arginine infused granulation tissue thickness compared with controls between days 4 and 14 (P < 0.05). Wound vessel count and luminal vascular surface area estimates derived from image analysis of histological sections were two- to three-fold lower in the L-arginine animals compared with controls (P < 0.05). Significant and sustained increases in wound fluid NOx levels were noted in L-arginine animals compared to saline controls (230 microM versus 75 microM at day 14, P < 0.05). Conversely, late VEGF levels (days 11 to 14) were reduced in the L-arginine animals compared to controls (7500 pg/ml versus 10,000 pg/ml at day 11, P < 0.05; 7250 pg/ml versus 11,101 pg/ml at day 14, P < 0.05). Arginine concentrations remained two- to four-fold greater in L-arginine treated animals compared with controls over the entire time course (P < 0.05). There were no significant differences in concentrations of ornithine, citrulline, or proline noted between groups over the 14-day period. Finally, TGF-beta1 levels were unaffected by L-arginine treatment. CONCLUSION Although NO appears to be necessary for granulation tissue formation, early supplemental arginine may disturb the reciprocal regulation of NOS 2 and arginase, leading to the preferential metabolism of arginine to excess NO rather than ornithine, with consequent reductions in angiogenesis and granulation tissue formation.
Collapse
Affiliation(s)
- Daithi Heffernan
- Department of Surgery, University of New Mexico HSC, Albuquerque, New Mexico 87131-0001, USA
| | | | | | | |
Collapse
|