1
|
Jin H, Wu Y, Zhang C, Zheng R, Xu H, Yang J, Li L. Tranilast alleviates skin inflammation and fibrosis in rosacea-like mice induced by long-term exposure to LL-37. Biochem Biophys Res Commun 2024; 737:150523. [PMID: 39133985 DOI: 10.1016/j.bbrc.2024.150523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/29/2024] [Accepted: 08/07/2024] [Indexed: 11/13/2024]
Abstract
Rosacea, a prevalent chronic facial inflammatory condition, afflicts millions worldwide. Its multifaceted pathogenesis poses challenges for effective treatment. Tranilast (TR), an analog of a tryptophan metabolite, has demonstrated anti-inflammatory and anti-fibrotic properties across various diseases. Yet, its potential in rosacea treatment remains understudied. Here, we induced rosacea-like symptoms in mice via prolonged LL-37 injections and administered TR intervention. Our findings reveal that TR mitigated skin lesions, reduced skin thickness, and suppressed inflammatory cell infiltration within the dermis of LL-37 mice. Notably, TR downregulated the expression of rosacea-associated inflammatory cytokines (TNF-α, IL-6, IL-1β, and IL-18) and the antimicrobial peptide CAMP, while also inhibiting NLRP3 inflammasome activation and the TLR4 signaling pathway. Furthermore, TR attenuated LL-37-induced fibrosis and hindered the transforming growth factor-β1 (TGF-β1)/Smad2/3 pathway. In summary, our study underscores TR's therapeutic potential in rosacea by mitigating both skin inflammation and fibrosis, thereby offering a promising treatment avenue for this condition.
Collapse
Affiliation(s)
- Hui Jin
- Department of Dermatology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100000, China; Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, China
| | - Yiling Wu
- Clinical Medical College, North China University of Science and Technology, Tangshan, 063210, China
| | - Chuanxi Zhang
- Clinical Medical College, North China University of Science and Technology, Tangshan, 063210, China
| | - Ruiping Zheng
- Clinical Medical College, North China University of Science and Technology, Tangshan, 063210, China
| | - Hong Xu
- Health Science Center, North China University of Science and Technology, Tangshan, 063210, China
| | - Jie Yang
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, China.
| | - Linfeng Li
- Department of Dermatology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100000, China.
| |
Collapse
|
2
|
Massoud G, Parish M, Hazimeh D, Moukarzel P, Singh B, Cayton Vaught KC, Segars J, Islam MS. Unlocking the potential of tranilast: Targeting fibrotic signaling pathways for therapeutic benefit. Int Immunopharmacol 2024; 137:112423. [PMID: 38861914 PMCID: PMC11245748 DOI: 10.1016/j.intimp.2024.112423] [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: 01/03/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Fibrosis is the excessive deposition of extracellular matrix in an organ or tissue that results from an impaired tissue repair in response to tissue injury or chronic inflammation. The progressive nature of fibrotic diseases and limited treatment options represent significant healthcare challenges. Despite the substantial progress in understanding the mechanisms of fibrosis, a gap persists translating this knowledge into effective therapeutics. Here, we discuss the critical mediators involved in fibrosis and the role of tranilast as a potential antifibrotic drug to treat fibrotic conditions. Tranilast, an antiallergy drug, is a derivative of tryptophan and has been studied for its role in various fibrotic diseases. These include scleroderma, keloid and hypertrophic scars, liver fibrosis, renal fibrosis, cardiac fibrosis, pulmonary fibrosis, and uterine fibroids. Tranilast exerts antifibrotic effects by suppressing fibrotic pathways, including TGF-β, and MPAK. Because it disrupts fibrotic pathways and has demonstrated beneficial effects against keloid and hypertrophic scars, tranilast could be used to treat other conditions characterized by fibrosis.
Collapse
Affiliation(s)
- Gaelle Massoud
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Maclaine Parish
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Dana Hazimeh
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Pamela Moukarzel
- American University of Beirut Medical Center, Faculty of Medicine, Riad El Solh, Beirut, Lebanon
| | - Bhuchitra Singh
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Kamaria C Cayton Vaught
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - James Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA.
| | - Md Soriful Islam
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
3
|
Saito H, Fushida S, Harada S, Miyashita T, Oyama K, Yamaguchi T, Tsukada T, Kinoshita J, Tajima H, Ninomiya I, Ohta T. Importance of human peritoneal mesothelial cells in the progression, fibrosis, and control of gastric cancer: inhibition of growth and fibrosis by tranilast. Gastric Cancer 2018; 21:55-67. [PMID: 28540637 PMCID: PMC5741788 DOI: 10.1007/s10120-017-0726-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Scirrhous gastric cancer is an intractable disease with a high incidence of peritoneal dissemination and obstructive symptoms (e.g., ileus, jaundice, and hydronephrosis) arising from accompanying marked fibrosis. Microenvironmental interactions between cancer cells and cancer-associated fibroblasts are the suggested cause of the disease. We elucidated the mechanisms of tumor growth and fibrosis using human peritoneal mesothelial cells (HPMCs) and investigated the effects of tranilast treatment on cells and a xenograft mouse model of fibrosis. METHODS HPMCs were isolated from surgically excised omentum and their interaction with MKN-45 gastric cancer cells was investigated using co-culture. Furthermore, a fibrosis tumor model was developed based on subcutaneous transplantation of co-cultured cells into the dorsal side of nude mice to form large fibrotic tumors. Mice were subsequently treated with or without tranilast. RESULTS The morphology of HPMCs treated with transforming growth factor (TGF)-β1 changed from cobblestone to spindle-type. Moreover, E-cadherin was weakly expressed whereas high levels of α-smooth muscle actin expression were observed. TGF-β-mediated epithelial-mesenchymal transition-like changes in HPMCs were inhibited in a dose-dependent manner following tranilast treatment through inhibition of Smad2 phosphorylation. In the mouse model, tumor size decreased significantly and fibrosis was inhibited in the tranilast treatment group compared with that in the control group. CONCLUSIONS Tranilast acts on the TGF-β/Smad pathway to inhibit interactions between cancer cells and cancer-associated fibroblasts, thereby inhibiting tumor growth and fibrosis. This study supports the hypothesis that tranilast represents a novel strategy to prevent fibrous tumor establishment represented by peritoneal dissemination.
Collapse
Affiliation(s)
- Hiroto Saito
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa 920-8641 Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Katsunobu Oyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Takahisa Yamaguchi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Tomoya Tsukada
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641 Japan
| |
Collapse
|
4
|
Genomic correlates of glatiramer acetate adverse cardiovascular effects lead to a novel locus mediating coronary risk. PLoS One 2017; 12:e0182999. [PMID: 28829817 PMCID: PMC5567477 DOI: 10.1371/journal.pone.0182999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022] Open
Abstract
Glatiramer acetate is used therapeutically in multiple sclerosis but also known for adverse effects including elevated coronary artery disease (CAD) risk. The mechanisms underlying the cardiovascular side effects of the medication are unclear. Here, we made use of the chromosomal variation in the genes that are known to be affected by glatiramer treatment. Focusing on genes and gene products reported by drug-gene interaction database to interact with glatiramer acetate we explored a large meta-analysis on CAD genome-wide association studies aiming firstly, to investigate whether variants in these genes also affect cardiovascular risk and secondly, to identify new CAD risk genes. We traced association signals in a 200-kb region around genomic positions of genes interacting with glatiramer in up to 60 801 CAD cases and 123 504 controls. We validated the identified association in additional 21 934 CAD cases and 76 087 controls. We identified three new CAD risk alleles within the TGFB1 region on chromosome 19 that independently affect CAD risk. The lead SNP rs12459996 was genome-wide significantly associated with CAD in the extended meta-analysis (odds ratio 1.09, p = 1.58×10−12). The other two SNPs at the locus were not in linkage disequilibrium with the lead SNP and by a conditional analysis showed p-values of 4.05 × 10−10 and 2.21 × 10−6. Thus, studying genes reported to interact with glatiramer acetate we identified genetic variants that concordantly with the drug increase the risk of CAD. Of these, TGFB1 displayed signal for association. Indeed, the gene has been associated with CAD previously in both in vivo and in vitro studies. Here we establish genome-wide significant association with CAD in large human samples.
Collapse
|
5
|
Zhang H, Ren KF, Chang H, Wang JL, Ji J. Surface-mediated transfection of a pDNA vector encoding short hairpin RNA to downregulate TGF-β1 expression for the prevention of in-stent restenosis. Biomaterials 2017; 116:95-105. [DOI: 10.1016/j.biomaterials.2016.11.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/28/2016] [Accepted: 11/24/2016] [Indexed: 01/14/2023]
|
6
|
Abstract
The authors discuss the mechanism of action, clinical trial data, and economic impact of both the paclitaxel and sirolimus drug eluting stents (DESs). Both DESs have been approved by the Food and Drug Administration for the treatment of native coronary arteries to prevent in-stent restenosis (ISR), which patients have experienced since the advent of balloon angioplasty and the bare metal stent. In-stent restenosis, which manifests itself as ischemic symptoms in patients, occurs as a result of the healing process after stent implantation. Until now, there has not been an effective method to prevent ISR. The sirolimus and paclitaxel DESs elute agents that act locally by different mechanisms to reduce neointimal hyperplasia, which is primarily responsible for ISR. Both DESs are capable of reducing the rate of ISR. There are certain physical and mechanistic differences between the 2 stents; the stents have not been compared head to head. Currently, they are indicated for uncomplicated native coronary lesions. Further investigation is needed to define their roles in the treatment of more complex lesions.
Collapse
Affiliation(s)
- William Alvarez
- The Johns Hopkins Hospital, Department of Pharmacy, 600 North Wolfe Street, Carnegie 180, Baltimore, MD 21287, The Johns Hopkins Hospital, Baltimore, Maryland
| | | |
Collapse
|
7
|
Wang H, Weihrauch D, Kersten JR, Toth JM, Passerini AG, Rajamani A, Schrepfer S, LaDisa JF. Alagebrium inhibits neointimal hyperplasia and restores distributions of wall shear stress by reducing downstream vascular resistance in obese and diabetic rats. Am J Physiol Heart Circ Physiol 2015; 309:H1130-40. [PMID: 26254329 DOI: 10.1152/ajpheart.00123.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 08/03/2015] [Indexed: 01/28/2023]
Abstract
Mechanisms of restenosis in type 2 diabetes mellitus (T2DM) are incompletely elucidated, but advanced glycation end-product (AGE)-induced vascular remodeling likely contributes. We tested the hypothesis that AGE-related collagen cross-linking (ARCC) leads to increased downstream vascular resistance and altered in-stent hemodynamics, thereby promoting neointimal hyperplasia (NH) in T2DM. We proposed that decreasing ARCC with ALT-711 (Alagebrium) would mitigate this response. Abdominal aortic stents were implanted in Zucker lean (ZL), obese (ZO), and diabetic (ZD) rats. Blood flow, vessel diameter, and wall shear stress (WSS) were calculated after 21 days, and NH was quantified. Arterial segments (aorta, carotid, iliac, femoral, and arterioles) were harvested to detect ARCC and protein expression, including transforming growth factor-β (TGF-β) and receptor for AGEs (RAGE). Downstream resistance was elevated (60%), whereas flow and WSS were significantly decreased (44% and 56%) in ZD vs. ZL rats. NH was increased in ZO but not ZD rats. ALT-711 reduced ARCC and resistance (46%) in ZD rats while decreasing NH and producing similar in-stent WSS across groups. No consistent differences in RAGE or TGF-β expression were observed in arterial segments. ALT-711 modified lectin-type oxidized LDL receptor 1 but not RAGE expression by cells on decellularized matrices. In conclusion, ALT-711 decreased ARCC, increased in-stent flow rate, and reduced NH in ZO and ZD rats through RAGE-independent pathways. The study supports an important role for AGE-induced remodeling within and downstream of stent implantation to promote enhanced NH in T2DM.
Collapse
Affiliation(s)
- Hongfeng Wang
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin
| | - Dorothee Weihrauch
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Judy R Kersten
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeffrey M Toth
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin; Department of Orthopaedic Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Anita Rajamani
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Sonja Schrepfer
- Transplant and Stem Cell Immunobiology Laboratory, University Heart Center and Cardiovascular Research Center, University of Hamburg, Hamburg, Germany; Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California
| | - John F LaDisa
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin; Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
8
|
Darakhshan S, Pour AB. Tranilast: a review of its therapeutic applications. Pharmacol Res 2014; 91:15-28. [PMID: 25447595 DOI: 10.1016/j.phrs.2014.10.009] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 12/14/2022]
Abstract
Tranilast (N-[3',4'-dimethoxycinnamoyl]-anthranilic acid) is an analog of a tryptophan metabolite. Initially, tranilast was identified as an anti-allergic agent, and used in the treatment of inflammatory diseases, such as bronchial asthma, atypical dermatitis, allergic conjunctivitis, keloids and hypertrophic scars. Subsequently, the results showed that it could be also effective in the management of a wide range of conditions. The beneficial effects of tranilast have also been seen in a variety of disease states, such as fibrosis, proliferative disorders, cancer, cardiovascular problems, autoimmune disorders, ocular diseases, diabetes and renal diseases. Moreover, several trials have shown that it has very low adverse effects and it is generally well tolerated by patients. In this review, we have attempted to accurately summarize previously published studies relating to the use of tranilast for a range of disorders and discuss the drug's possible mode of action. The major mode of the drug's efficacy appears to be the suppression of the expression and/or action of the TGF-β pathway, but the drug affects other factors as well. The findings presented in this review demonstrate the potential of tranilast for the control of a vast array of pathological situations, furthermore, it is a prescribed drug without severe side effects.
Collapse
Affiliation(s)
- Sara Darakhshan
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Ali Bidmeshki Pour
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.
| |
Collapse
|
9
|
Xu J, Shi GP. Vascular wall extracellular matrix proteins and vascular diseases. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2106-2119. [PMID: 25045854 DOI: 10.1016/j.bbadis.2014.07.008] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 07/07/2014] [Accepted: 07/14/2014] [Indexed: 01/08/2023]
Abstract
Extracellular matrix proteins form the basic structure of blood vessels. Along with providing basic structural support to blood vessels, matrix proteins interact with different sets of vascular cells via cell surface integrin or non-integrin receptors. Such interactions induce vascular cell de novo synthesis of new matrix proteins during blood vessel development or remodeling. Under pathological conditions, vascular matrix proteins undergo proteolytic processing, yielding bioactive fragments to influence vascular wall matrix remodeling. Vascular cells also produce alternatively spliced variants that induce vascular cell production of different matrix proteins to interrupt matrix homeostasis, leading to increased blood vessel stiffness; vascular cell migration, proliferation, or death; or vascular wall leakage and rupture. Destruction of vascular matrix proteins leads to vascular cell or blood-borne leukocyte accumulation, proliferation, and neointima formation within the vascular wall; blood vessels prone to uncontrolled enlargement during blood flow diastole; tortuous vein development; and neovascularization from existing pathological tissue microvessels. Here we summarize discoveries related to blood vessel matrix proteins within the past decade from basic and clinical studies in humans and animals - from expression to cross-linking, assembly, and degradation under physiological and vascular pathological conditions, including atherosclerosis, aortic aneurysms, varicose veins, and hypertension.
Collapse
Affiliation(s)
- Junyan Xu
- Department of Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Ostriker A, Horita HN, Poczobutt J, Weiser-Evans MCM, Nemenoff RA. Vascular smooth muscle cell-derived transforming growth factor-β promotes maturation of activated, neointima lesion-like macrophages. Arterioscler Thromb Vasc Biol 2014; 34:877-86. [PMID: 24526697 DOI: 10.1161/atvbaha.114.303214] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To define the contribution of vascular smooth muscle cell (SMC)-derived factors to macrophage phenotypic modulation in the setting of vascular injury. APPROACH AND RESULTS By flow cytometry, macrophages (M4) were the predominant myeloid cell type recruited to wire-injured femoral arteries, in mouse, compared with neutrophils or eosinophils. Recruited macrophages from injured vessels exhibited a distinct expression profile relative to circulating mononuclear cells (peripheral blood monocytes; increased: interleukin-6, interleukin-10, interleukin-12b, CC chemokine receptor [CCR]3, CCR7, tumor necrosis factor-α, inducible nitric oxide synthase, arginase 1; decreased: interleukin-12a, matrix metalloproteinase [MMP]9). This phenotype was recapitulated in vitro by maturing rat bone marrow cells in the presence of macrophage-colony stimulating factor and 20% conditioned media from cultured rat SMC (sMϕ) compared with maturation in macrophage-colony stimulating factor alone (M0). Recombinant transforming growth factor (TGF)-β1 recapitulated the effect of SMC conditioned media. Macrophage maturation studies performed in the presence of a pan-TGF-β neutralizing antibody, a TGF-β receptor inhibitor, or conditioned media from TGF-β-depleted SMCs confirmed that the SMC-derived factor responsible for macrophage activation was TGF-β. Finally, the effect of SMC-mediated macrophage activation on SMC biology was assessed. SMCs cocultured with sMϕ exhibited increased rates of proliferation relative to SMCs cultured alone or with M0 macrophages. CONCLUSIONS SMC-derived TGF-β modulates the phenotype of maturing macrophages in vitro, recapitulating the phenotype found in vascular lesions in vivo. SMC-modulated macrophages induce SMC activation to a greater extent than control macrophages.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- Cell Proliferation
- Cells, Cultured
- Coculture Techniques
- Culture Media, Conditioned/metabolism
- Cytokines/metabolism
- Disease Models, Animal
- Femoral Artery/injuries
- Femoral Artery/metabolism
- Femoral Artery/pathology
- Humans
- Macrophage Activation
- Macrophage Colony-Stimulating Factor/metabolism
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Paracrine Communication
- Phenotype
- RNA Interference
- Rats
- Time Factors
- Transfection
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta1/metabolism
- Vascular System Injuries/genetics
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Allison Ostriker
- From the Department of Medicine, Division of Renal Diseases and Hypertension (H.N.H., J.P., M.C.M.W.-E., R.A.N.), Department of Pharmacology (A.O., M.C.M.W.-E., R.A.N.), and Cardiovascular and Pulmonary Research Laboratory (M.C.M.W.-E., R.A.N.), University of Colorado Denver, Aurora
| | | | | | | | | |
Collapse
|
11
|
Role of TGF-β1/Smad3 signaling pathway in secretion of type I and III collagen by vascular smooth muscle cells of rats undergoing balloon injury. J Biomed Biotechnol 2012; 2012:965953. [PMID: 23091366 PMCID: PMC3471068 DOI: 10.1155/2012/965953] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 06/25/2012] [Accepted: 06/25/2012] [Indexed: 11/17/2022] Open
Abstract
Antisense Smad3 adenoviral vectors were used to transfect vascular smooth muscle cells (VSMCs) from rats with balloon injury or infused into the rat balloon-catheter injured carotid arteries, and the role of TGF-β1/Smad3 signaling pathway in the secretion of type I and III collagen by VSMCs following balloon injury was investigated. Antisense Smad3 adenoviral vectors were used to transfect these VSMCs (antisense Smad3 group). A total 90 rats were randomly assigned into blank control group, experiment group, negative control group. In the in vitro study, the expression of type I and III collagen was markedly reduced in the antisense Smad3 group when compared with the control groups (P < 0.05). In the in vivo study, the expression of type I and III collagen was significantly lower than that in the negative control group at 3 days, 1 week and 2 weeks after injury (P < 0.05). At 2 weeks and 3 months after injury, the lumen area in the antisense Smad3 group was markedly increased but the intimal area dramatically reduced when compared with the negative control (P < 0.05). We conclude that transfection of VSMCs with antisense Smad3 can reduce the secretion of type I and III collagen which then inhibit intimal hyperplasia.
Collapse
|
12
|
Wang CH, Lee YS, Lin SJ, Mei HF, Lin SY, Liu MH, Chen JR, Cherng WJ. Surface Markers of Heterogeneous Peripheral Blood–Derived Smooth Muscle Progenitor Cells. Arterioscler Thromb Vasc Biol 2012; 32:1875-83. [DOI: 10.1161/atvbaha.112.245852] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Chao-Hung Wang
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Yun-Shien Lee
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Shing-Jong Lin
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Hsiu-Fu Mei
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Sheng-Yuan Lin
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Min-Hui Liu
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Jim-Ray Chen
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| | - Wen-Jin Cherng
- From the Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan (C.-H.W., H.-F. M., S.-Y.L., M.-H.L., W.-J.C.); Department of Pathology, Chang Gung University College of Medicine, Taoyuan, Taiwan (J.-R.C.); Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan (Y.-S.L.); and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan (S.-J.L., C.-H.W.)
| |
Collapse
|
13
|
Sun DX, Liu Z, Tan XD, Cui DX, Wang BS, Dai XW. Nanoparticle-mediated local delivery of an antisense TGF-β1 construct inhibits intimal hyperplasia in autogenous vein grafts in rats. PLoS One 2012; 7:e41857. [PMID: 22860019 PMCID: PMC3408488 DOI: 10.1371/journal.pone.0041857] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 06/26/2012] [Indexed: 11/18/2022] Open
Abstract
Background Intimal hyperplasia is one of the most important causes of vascular graft failure. Numerous studies have correlated transforming growth factor-β1 (TGF-β1) with extracellular matrix (ECM) deposition, a hallmark of intimal thickening. Principal Findings In the present study, we performed immunohistochemistry, RT-PCR, and Western blot to examine the dynamic expression of TGF-β1, TGF-β1 receptor type I (TGF-β RI), matrix metalloproteinase-1 (MMP-1) and tissue inhibitor of metalloproteinase-1 (TIMP-1) during intimal hyperplasia in grafted veins of a rat model generated by grafting a portion of the right internal jugular vein to the ipisiliary caroid artery. Additionally, we determined whether nanoparticle-mediated delivery of a TGF-β1 antisense-expressing construct prevented TGF-β1 expression and intimal hyperplasia in grafted veins. In grafted veins, the expression of TGF-β1 significantly increased on day 3 after transplantation, peaked on day 7, slightly decreased on day 14, and returned to baseline levels on day 28. The positive expression of TGF-β RI in grafted veins remarkably increased on day 7, peaked on day 14, and decreased thereafter. MMP-1 expression decreased significantly, while TIMP-1 expression increased, significantly on days 14 and 28. Nanoparticle-mediated delivery of a TGF-β1 antisense-expressing construct down-regulated TGF-β1 expression and inhibited intimal hyperplasia in grafted veins. Conclusions Our findings provide further evidence that TGF-β1 plays an integral role in the development of intimal hyperplasia after vascular injury. Nanoparticle-mediated delivery of a TGF-β1 antisense-expressing construct is a feasible strategy to target TGF-β1-induced intimal thickening.
Collapse
Affiliation(s)
- Da-Xin Sun
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
- * E-mail: (XWD); (DXS)
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Dong-Xu Cui
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Bao-Sheng Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xian-Wei Dai
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
- * E-mail: (XWD); (DXS)
| |
Collapse
|
14
|
Osherov AB, Gotha L, Cheema AN, Qiang B, Strauss BH. Proteins mediating collagen biosynthesis and accumulation in arterial repair: novel targets for anti-restenosis therapy. Cardiovasc Res 2011; 91:16-26. [PMID: 21245059 DOI: 10.1093/cvr/cvr012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Events contributing to restenosis after coronary interventions include platelet aggregation, inflammatory cell infiltration, growth factor release, and accumulation of smooth muscle cells (SMCs) and extracellular matrix (ECM). The ECM is composed of various collagen subtypes and proteoglycans and over time constitutes the major component of the mature restenotic plaque. The pathophysiology of collagen accumulation in the ECM during arterial restenosis is reviewed. Factors regulating collagen synthesis and degradation, including various cytokines and growth factors involved in the process, may be targets for therapies aimed at prevention of in-stent restenosis.
Collapse
Affiliation(s)
- Azriel B Osherov
- Schulich Heart Program, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Room A-253, Toronto, Ontario, Canada M4N 3M5
| | | | | | | | | |
Collapse
|
15
|
Braun A, Dofiles L, Rousselle S, Guerrero L, Gunther J, Yednock T, Stricker-Krongrad A, Messersmith E. Effects of an alpha-4 integrin inhibitor on restenosis in a new porcine model combining endothelial denudation and stent placement. PLoS One 2010; 5:e14314. [PMID: 21179441 PMCID: PMC3001476 DOI: 10.1371/journal.pone.0014314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 10/12/2010] [Indexed: 11/29/2022] Open
Abstract
Restenosis remains the main complication of balloon angioplasty and/or stent implantation. Preclinical testing of new pharmacologic agents preventing restenosis largely rely on porcine models, where restenosis is assessed after endothelial abrasion of the arterial wall or stent implantation. We combined endothelial cell denudation and implantation of stents to develop a new clinically relevant porcine model of restenosis, and used this model to determine the effects of an α4 integrin inhibitor, ELN 457946, on restenosis. Balloon-angioplasty endothelial cell denudation and subsequent implantation of bare metal stents in the left anterior descending coronary, iliac, and left common carotid arteries was performed in domestic pigs, treated with vehicle or ELN 457946, once weekly via subcutaneous injections, for four weeks. After 1 month, histopathology and morphometric analyses of the arteries showed complete healing and robust, consistent restenotic response in stented arteries. Treatment with ELN 457946 resulted in a reduction in the neointimal response, with decreases in area percent stenosis between 12% in coronary arteries and 30% in peripheral vessels. This is the first description of a successful pig model combining endothelial cell denudation and bare metal stent implantation. This new double injury model may prove particularly useful to assess pharmacological effects of drug candidates on restenosis, in coronary and/or peripheral arteries. Furthermore, the ELN 457946 α4 integrin inhibitor, administered subcutaneously, reduced inflammation and restenosis in stented coronary and peripheral arteries in pigs, therefore representing a promising systemic therapeutic approach in reducing restenosis in patients undergoing angioplasty and/or stent implantation.
Collapse
Affiliation(s)
- Anne Braun
- Charles River Laboratories, Preclinical Services Massachusetts, Shrewsbury, Massachusetts, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Westerweel PE, van Velthoven CTJ, Nguyen TQ, den Ouden K, de Kleijn DPV, Goumans MJ, Goldschmeding R, Verhaar MC. Modulation of TGF-β/BMP-6 expression and increased levels of circulating smooth muscle progenitor cells in a type I diabetes mouse model. Cardiovasc Diabetol 2010; 9:55. [PMID: 20858224 PMCID: PMC2954908 DOI: 10.1186/1475-2840-9-55] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 09/21/2010] [Indexed: 12/03/2022] Open
Abstract
Background Diabetic patients experience exaggerated intimal hyperplasia after endovascular procedures. Recently it has been shown that circulating smooth muscle progenitor cells (SPC) contribute to intimal hyperplasia. We hypothesized that SPC differentiation would be increased in diabetes and focused on modulation of TGF-β/BMP-6 signaling as potential underlying mechanism. Methods We isolated SPC from C57Bl/6 mice with streptozotocin-induced diabetes and controls. SPC differentiation was evaluated by immunofluorescent staining for αSMA and collagen Type I. SPC mRNA expression of TGF-β and BMP-6 was quantified using real-time PCR. Intima formation was assessed in cuffed femoral arteries. Homing of bone marrow derived cells to cuffed arterial segments was evaluated in animals transplanted with bone marrow from GFP-transgenic mice. Results We observed that SPC differentiation was accelerated and numeric outgrowth increased in diabetic animals (24.6 ± 8.8 vs 8.3 ± 1.9 per HPF after 10 days, p < 0.05). Quantitative real-time PCR showed increased expression of TGF-β and decreased expression of the BMP-6 in diabetic SPC. SPC were MAC-3 positive, indicative of monocytic lineage. Intima formation in cuffed arterial segments was increased in diabetic mice (intima/media ratio 0.68 ± 0.15 vs 0.29 ± 0.06, p < 0.05). In GFP-chimeric mice, bone marrow derived cells were observed in the neointima (4.4 ± 3.3 cells per section) and particularly in the adventitia (43.6 ± 9.3 cells per section). GFP-positive cells were in part MAC-3 positive, but rarely expressed α-SMA. Conclusions In conclusion, in a diabetic mouse model, SPC levels are increased and SPC TGF-β/BMP-6 expression is modulated. Altered TGF-β/BMP-6 expression is known to regulate smooth muscle cell differentiation and may facilitate SPC differentiation. This may contribute to exaggerated intimal hyperplasia in diabetes as bone marrow derived cells home to sites of neointima formation.
Collapse
Affiliation(s)
- Peter E Westerweel
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Saegusa Y, Sadakane C, Koseki J, Hasegawa Y, Shindo S, Maruyama H, Kase Y, Takeda S, Takeda H, Hattori T. Effect of TJN-331 on Anti-Thy1 Nephritis in Rats via Inhibition of Transforming Growth Factor-.BETA.1 Production. Biol Pharm Bull 2010; 33:1710-5. [DOI: 10.1248/bpb.33.1710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yayoi Saegusa
- Department of Pathophysiology and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University
- Tsumura & Co., Tsumura Research Laboratories
| | - Chiharu Sadakane
- Department of Pathophysiology and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University
- Tsumura & Co., Tsumura Research Laboratories
| | | | | | | | | | - Yoshio Kase
- Tsumura & Co., Tsumura Research Laboratories
| | | | - Hiroshi Takeda
- Department of Pathophysiology and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University
| | | |
Collapse
|
18
|
Pallero MA, Talbert Roden M, Chen YF, Anderson PG, Lemons J, Brott BC, Murphy-Ullrich JE. Stainless steel ions stimulate increased thrombospondin-1-dependent TGF-beta activation by vascular smooth muscle cells: implications for in-stent restenosis. J Vasc Res 2009; 47:309-22. [PMID: 20016205 DOI: 10.1159/000265565] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Accepted: 06/02/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Despite advances in stent design, in-stent restenosis (ISR) remains a significant clinical problem. All implant metals exhibit corrosion, which results in release of metal ions. Stainless steel (SS), a metal alloy widely used in stents, releases ions to the vessel wall and induces reactive oxygen species, inflammation and fibroproliferative responses. The molecular mechanisms are unknown. TGF-beta is known to be involved in the fibroproliferative responses of vascular smooth muscle cells (VSMCs) in restenosis, and TGF-beta antagonists attenuate ISR. We hypothesized that SS ions induce the latent TGF-beta activator, thrombospondin-1 (TSP1), through altered oxidative signaling to stimulate increased TGF-beta activation and VSMC phenotype change. METHODS VSMCs were treated with SS metal ion cocktails, and morphology, TSP1, extracellular matrix production, desmin and TGF-beta activity were assessed by immunoblotting. RESULTS SS ions stimulate the synthetic phenotype, increased TGF-beta activity, TSP1, increased extracellular matrix and downregulation of desmin in VSMCs. Furthermore, SS ions increase hydrogen peroxide and decrease cGMP-dependent protein kinase (PKG) signaling, a known repressor of TSP1 transcription. Catalase blocks SS ion attenuation of PKG signaling and increased TSP1 expression. CONCLUSIONS These data suggest that ions from stent alloy corrosion contribute to ISR through stimulation of TSP1-dependent TGF-beta activation.
Collapse
Affiliation(s)
- Manuel A Pallero
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Lee-Manion AM, Price RK, Strain JJ, Dimberg LH, Sunnerheim K, Welch RW. In vitro antioxidant activity and antigenotoxic effects of avenanthramides and related compounds. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2009; 57:10619-10624. [PMID: 19874025 DOI: 10.1021/jf9024739] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Avenanthramides are substituted N-cinnamoylanthranilic acids, with hydroxycinnamic acid and anthranilic acid moieties. These alkaloid phenols, which are unique to oats, may confer health benefits via antioxidant or other mechanisms. Synthetic avenanthramides, hydroxycinnamic acids, Tranilast, and ascorbic acid were evaluated for antioxidant activity using two assays, DPPH (2,2-diphenyl-1-picrylhydrazyl) and FRAP (ferric reducing antioxidant potential), and for antigenotoxicity using the Comet assay with stressed human adenocarcinoma colon cells. Of all the compounds tested, N-(3',4'-dihydroxy-(E)-cinnamoyl)-5-hydroxyanthranilic acid (2c), an abundant oat avenanthramide, generally had the highest activity in all three assays. The drug Tranilast showed antigenotoxic effects, but not antioxidant activity, suggesting that antigenotoxicity is not dependent on antioxidant effects. Overall, results show that avenanthramides exert antioxidant and antigenotoxic activities that are comparable to those of ascorbic acid and which have the potential to exert beneficial physiological effects.
Collapse
Affiliation(s)
- Alison M Lee-Manion
- Northern Ireland Centre for Food and Health, School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, United Kingdom
| | | | | | | | | | | |
Collapse
|
20
|
Yao EH, Fukuda N, Ueno T, Matsuda H, Nagase H, Matsumoto Y, Sugiyama H, Matsumoto K. A pyrrole-imidazole polyamide targeting transforming growth factor-beta1 inhibits restenosis and preserves endothelialization in the injured artery. Cardiovasc Res 2008; 81:797-804. [PMID: 19098300 DOI: 10.1093/cvr/cvn355] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Although the use of drug-eluting stents (DESs) has been shown to limit neointima hyperplasia, currently available DESs may adversely affect re-endothelialization. To evaluate whether a novel gene silencer pyrrole-imidazole (PI) polyamide targeting transforming growth factor (TGF)-beta1 is a candidate agent for the DESs, we examined the effects of PI polyamide targeting the TGF-beta1 promoter on neointimal formation in rat carotid artery after balloon injury. METHODS AND RESULTS PI polyamide was designed to span the boundary of the AP-1 binding site of the TGF-beta1 promoter. After inducing balloon injury to arteries, incubation with PI polyamide was carried out for 10 min. Neointimal thickening and re-endothelialization were evaluated at 21 days after injury. Fluoresceinisothiocyanate-labelled PI polyamide was distributed into most of the nuclei in the injured artery without any delivery reagents. PI polyamide (100 microg) significantly inhibited neointimal thickening at 21 days after injury by 57%. PI polyamide targeting TGF-beta1 significantly decreased the expression of TGF-beta1 mRNA and protein in the artery at 3 days after injury and also suppressed the expression of connective tissue growth factor (CTGF), fibronectin, collagen type 1, and lectin-like ox-LDL receptor-1 mRNAs. A morphometric analysis showed that PI polyamide targeting TGF-beta1 accelerated re-endothelialization in the injured artery. CONCLUSION These findings suggest that the synthetic PI polyamide targeting the TGF-beta1 promoter may have the potential to suppress neointimal hyperplasia after arterial injury by the down-regulation of TGF-beta1 and CTGF and the reduction of the extracellular matrix. As a result, PI polyamide targeting TGF-beta1 may therefore be a potentially effective agent for the treatment of in-stent restenosis, as a candidate agent for the next-generation DES.
Collapse
Affiliation(s)
- En-Hui Yao
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Gordon KJ, Blobe GC. Role of transforming growth factor-beta superfamily signaling pathways in human disease. Biochim Biophys Acta Mol Basis Dis 2008; 1782:197-228. [PMID: 18313409 DOI: 10.1016/j.bbadis.2008.01.006] [Citation(s) in RCA: 490] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 01/22/2008] [Accepted: 01/23/2008] [Indexed: 12/14/2022]
Abstract
Transforming growth factor beta (TGF-beta) superfamily signaling pathways are ubiquitous and essential regulators of cellular processes including proliferation, differentiation, migration, and survival, as well as physiological processes, including embryonic development, angiogenesis, and wound healing. Alterations in these pathways, including either germ-line or somatic mutations or alterations in the expression of members of these signaling pathways often result in human disease. Appropriate regulation of these pathways is required at all levels, particularly at the ligand level, with either a deficiency or an excess of specific TGF-beta superfamily ligands resulting in human disease. TGF-beta superfamily ligands and members of these TGF-beta superfamily signaling pathways also have emerging roles as diagnostic, prognostic or predictive markers for human disease. Ongoing studies will enable targeting of TGF-beta superfamily signaling pathways for the chemoprevention and treatment of human disease.
Collapse
Affiliation(s)
- Kelly J Gordon
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | | |
Collapse
|
22
|
Tuleta I, Skowasch D, Peuster M, Nickenig G, Bauriedel G. Cells of primarily extravascular origin in neointima formation following stent implantation: coordinated expression of endothelial progenitor, dendritic and neural crest-derived cells. Cardiology 2007; 110:199-205. [PMID: 18057885 DOI: 10.1159/000111930] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Accepted: 07/13/2007] [Indexed: 11/19/2022]
Abstract
OBJECTIVES In-stent restenosis due to neointima formation is a major limitation following stent implantation. Recently, several studies reported mobilization of primarily extravascular cells to the arterial sites after balloon angioplasty. Therefore, the goal of the present study was to assess the coordinated neointimal expression of endothelial progenitor, dendritic and neural crest-derived cells after stent implantation. METHODS Male minipigs underwent stent implantation in abdominal aortic segments. Animals were sacrificed at 1, 7, 14, 30, 60 or 90 days. Cross sections of the injured vessels were obtained for immunohistochemistry using specific antibodies for the detection of endothelial progenitor (CD133), dendritic (S100) and neural crest-derived cells (GFAP), as well as monocytes/macrophages (CD14) and T lymphocytes (CD3). RESULTS As a key finding, frequency of CD133, S100, GFAP, CD14 and CD3 (18.5 +/- 3.6, 14.9 +/- 1.8, 10.6 +/- 1.1, 40.2 +/- 8.3 and 5.0 +/- 0.6%, respectively) in neointima was maximal at day 7. With ongoing neointima enlargement, expression of these cells decreased. In advanced neointima, labeled cells were predominantly localized at luminal and stented sites. Media showed almost no immunoreactivity of the markers studied, whereas adventitial zones of neovascularization revealed some signals. CONCLUSIONS Endothelial progenitor, dendritic, neural crest-derived and inflammatory cells are consistently recruited into arterial neointima, mostly at early time points after stent implantation.
Collapse
Affiliation(s)
- Izabela Tuleta
- Department of Internal Medicine II - Cardiology, University of Bonn, Bonn, Germany
| | | | | | | | | |
Collapse
|
23
|
Prud'homme GJ. Pathobiology of transforming growth factor beta in cancer, fibrosis and immunologic disease, and therapeutic considerations. J Transl Med 2007; 87:1077-91. [PMID: 17724448 DOI: 10.1038/labinvest.3700669] [Citation(s) in RCA: 311] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) is a highly pleiotropic cytokine that plays an important role in wound healing, angiogenesis, immunoregulation and cancer. The cells of the immune system produce the TGF-beta1 isoform, which exerts powerful anti-inflammatory functions, and is a master regulator of the immune response. However, this is context dependent, because TGF-beta can contribute to the differentiation of both regulatory (suppressive) T cells (Tr cells) and inflammatory Th17 cells. While TGF-beta might be underproduced in some autoimmune diseases, it is overproduced in many pathological conditions. This includes pulmonary fibrosis, glomerulosclerosis, renal interstitial fibrosis, cirrhosis, Crohn's disease, cardiomyopathy, scleroderma and chronic graft-vs-host disease. In neoplastic disease, TGF-beta suppresses the progression of early lesions, but later this effect is lost and cancer cells produce TGF-beta, which then promotes metastasis. This cytokine also contributes to the formation of the tumor stroma, angiogenesis and immunosuppression. In view of this, several approaches are being studied to inhibit TGF-beta activity, including neutralizing antibodies, soluble receptors, receptor kinase antagonist drugs, antisense reagents and a number of less specific drugs such as angiotensin II antagonists and tranilast. It might be assumed that TGF-beta blockade would result in severe inflammatory disease, but this has not been the case, presumably because the neutralization is only partial. In contrast, the systemic administration of TGF-beta for therapeutic purposes is limited by toxicity and safety concerns, but local administration appears feasible, especially to promote wound healing. Immunotherapy or vaccination stimulating TGF-beta production and/or Tr differentiation might be applied to the treatment of autoimmune diseases. The benefits of new therapies targeting TGF-beta are under intense investigation.
Collapse
Affiliation(s)
- Gérald J Prud'homme
- Department of Laboratory Medicine, St Michael's Hospital and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Karakayali F, Haberal N, Tufan H, Hasirci N, Basaran O, Sevmis S, Akdur A, Kiziltay A, Haberal M. Evaluation of neointimal hyperplasia on tranilast-coated synthetic vascular grafts: an experimental study. J INVEST SURG 2007; 20:167-73. [PMID: 17613691 DOI: 10.1080/08941930701365101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Tranilast is an antiallergic drug that interferes with proliferation and migration of vascular smooth muscle cell induced by platelet-derived growth factor (PDGF) and transforming growth factor-beta1 (TGF-beta1). We investigated the local effect of tranilast on neointimal hyperplasia using tranilast-coated prosthetic grafts. The inner sides of the thin-walled polytetrafluoroethylene (PTFE) grafts were coated with chitosan and tranilast containing chitosan solution. Wistar albino rats (32) were used in the study. Patches (1 x 2 mm) for vascular grafts were prepared. Three groups were tested: group 1 (n = 12; tranilast coated), group 2 (n = 10; adhesive-only film-layer-coated), and group 3 (n = 10; normal ePTFE patch grafts sutured to the carotid arteries of the rats). Recipient sites of the carotid arteries were excised 4 weeks after surgery. All sections were examined histologically for graft patency, thrombus formation, and neointimal thickness. Expression of PDGF, fibroblast growth factor, and TGF-beta1 on cross-sections of the neointima were evaluated by immunohistochemistry. No significant differences were found regarding mean neointimal thicknesses. PDGF and TGF-beta-1 expressions were significantly lower in group 1. Although a decrease in local effect of tranilast was observed for growth factor expressions at a drug concentration of 0.05 mg/cm(2), a significant reduction in neointimal hyperplasia was not achieved. The coating concentration of 0.05 mg/cm(2) may have been too low to produce an antiproliferative effect. Given our promising results, further studies are recommended and planned using different drug concentrations and time intervals.
Collapse
Affiliation(s)
- Feza Karakayali
- Department of General Surgery, Baskent University Faculty of Medicine, Ankara, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wildgruber M, Weiss W, Berger H, Wolf O, Eckstein HH, Heider P. Association of Circulating Transforming Growth Factor beta, Tumor Necrosis Factor alpha and Basic Fibroblast Growth Factor with Restenosis after Transluminal Angioplasty. Eur J Vasc Endovasc Surg 2007; 34:35-43. [PMID: 17475518 DOI: 10.1016/j.ejvs.2007.02.009] [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] [Received: 11/01/2006] [Accepted: 02/15/2007] [Indexed: 11/23/2022]
Abstract
OBJECTIVES To assess prospectively the early time course of Transforming Growth Factor beta-1 (TGFbeta-1), basic Fibroblast Growth Factor (bFGF) and Tumor Necrosis Factor alpha (TNFalpha) as possible contributors to restenosis development after angioplasty. DESIGN Prospective Study. METHODS The levels of the soluble forms of these factors in the early response to Percutaneous Transluminal Angioplasty (PTA) in the arteries of the lower limb were prospectively assessed. 32 patients with peripheral arterial occlusive disease (PAOD), presenting with intermittent claudication (Fontaine stage IIb) were scheduled for angioplasty treatment. Serum levels of TGFbeta-1, TNFalpha and bFGF were assessed before intervention, 15 and 60 minutes after, 24 hours after as well as 2 and 4 weeks after intervention. We compared the distribution patterns between patients treated with balloon angioplasty and patients who required secondary stent implantation. Endpoint was the development of restenosis within 6 months after interventional treatment, defined as a lumen diameter reduction of more than 50% by ultrasound measurement compared to the result after PTA. RESULTS The patients who later developed restenosis had significantly higher levels of TGFbeta-1 at 15 minutes, 24 hours and 2 weeks after PTA (p<0.05). TNFalpha and bFGF were only detected in a few patients and no significant change of serum levels was observed. CONCLUSION The results demonstrate a possible role of TGFbeta-1 in the formation of restenosis after PTA.
Collapse
Affiliation(s)
- M Wildgruber
- Department of Vascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Cooper K, Young J, Wadsworth S, Cui H, diZerega GS, Rodgers KE. Reduction of post-surgical adhesion formation with tranilast. J Surg Res 2007; 141:153-61. [PMID: 17560610 DOI: 10.1016/j.jss.2006.05.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Revised: 05/05/2006] [Accepted: 05/10/2006] [Indexed: 11/29/2022]
Abstract
BACKGROUND Preclinical studies using the rabbit sidewall and double uterine horn models were used to assess time and dose response of tranilast delivered via subcutaneous pump, p.o., or as an intraperitoneal bolus in viscoelastic gels as well as an intraperitoneal biodegradable poly(p-dioxanone) fiber in reducing adhesions compared to vehicle controls. MATERIALS AND METHODS New Zealand white rabbits underwent laparotomy followed by: 1) uterine horn abrasion and peripheral devascularization or 2) cecal abrasion and sidewall deperitonealization. Tranilast treatment using various vehicles and dosages was compared to vehicle alone versus no treatment. Animals were euthanized after 7 to 21 days. Adhesion formation was assessed by two independent observers. RESULTS There were reductions in adhesion formation when drug was delivered topically, but oral drug alone was not effective. When tranilast was given preoperatively, oral drug added to the adhesion reduction of intraperitoneal administered drug. Tranilast in a viscoelastic carrier as well as in a biodegradable fiber was effective at reducing adhesions in the double uterine horn model. The slow release of tranilast from a biodegradable rod produced overall the best results. There were no safety issues. CONCLUSION Tranilast was effective in reducing adhesions when given in a variety of vehicles in different rabbit models of adhesion formation. Overall, the sustained intraperitoneal delivery of tranilast from biodegradable fibers was the most suitable for clinical testing.
Collapse
Affiliation(s)
- Kevin Cooper
- Center for Biomaterials & Advanced Technologies, Medical Devices Group, A division of Ethicon, A Johnson & Johnson Company, Somerville, New Jersey, USA
| | | | | | | | | | | |
Collapse
|
27
|
Joner M, Farb A, Cheng Q, Finn AV, Acampado E, Burke AP, Skorija K, Creighton W, Kolodgie FD, Gold HK, Virmani R. Pioglitazone Inhibits In-Stent Restenosis in Atherosclerotic Rabbits by Targeting Transforming Growth Factor-β and MCP-1. Arterioscler Thromb Vasc Biol 2007; 27:182-9. [PMID: 17068283 DOI: 10.1161/01.atv.0000251021.28725.e8] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Although emerging data from preclinical and clinical studies suggests a reduction of in-stent restenosis with peroxisome proliferator-activated receptor (PPAR)-γ agonists, the reduction of neointimal growth via anti-inflammatory mechanisms has not been explored.
Methods and Results—
Hypercholesterolemic New Zealand White rabbits (n=45) received bilateral balloon-expandable stents implanted into atherosclerotic iliac arteries. Animals were randomized to oral pioglitazone 3 (low dose) or 10 mg/kg per day (high dose) started on the day of stent implantation; control rabbits received placebo. Tissue harvest was performed 28 days after stenting, and stented segments underwent histology, morphometry, immunostaining for macrophages, and scanning electron microscopy. In selected animals, stented arterial segments were placed in organoid culture for 48 hours, and the conditioned media was assayed for 23 different cytokines. There was a 21% reduction in neointimal area for high-dose pioglitazone treated versus placebo rabbits (
P
<0.005), which was associated with a significant reduction of neointimal macrophages. Analysis of conditioned media revealed an 82% and 74% reduction in the release of monocyte chemoattractant protein-1 (MCP-1) (
P
<0.007) and transforming growth factor (TGF)-β1 (
P
<0.01), respectively, in stented segments from animals treated with 10 mg/kg per day pioglitazone versus placebo.
Conclusions—
Oral pioglitazone suppresses in-stent neointimal growth by limiting local inflammatory pathways and may be useful as an adjunctive therapy in patients undergoing percutaneous interventions.
Collapse
|
28
|
Charng MJ, Wu CH. Transcriptional activation of p21 by Tranilast is mediated via transforming growth factor beta signal pathway. Br J Pharmacol 2006; 147:117-24. [PMID: 16284627 PMCID: PMC1615847 DOI: 10.1038/sj.bjp.0706460] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Tranilast, an antiallergic medication, is a very promising inhibitor of restenosis after balloon angioplasty. Tranilast can prevent the proliferation and migration of smooth muscle cells by activating the gene expression of p21, a strong cyclin/cyclin-dependent kinase (CDK) inhibitor, and by arresting cell growth at the G0/G1 phase. The signaling pathway of Tranilast in regulating p21 is to our best interest and is elucidated in the present study. The major emphasis was weighted on exploring the regulatory effects of Tranilast on promoter activity of p21. By serial deletion analysis, the sequence between -74 and -83 bp of the p21 promoter, previously identified as the transforming growth factor-beta (TGF-beta)-response element, was found sufficient, where as most of the promoter region 5' to -111 bp was found unnecessary for the transcriptional activation of p21 by both TGF-beta1 and Tranilast. Tranilast was also found to induce phosphorylation of Smad2 (a cytoplasmic signaling molecule essential for mediating TGF-beta signal transduction). Transfection of DeltakTbetaRII, a truncated form of TGF-beta type II receptor known to exert a dominant-negative effect on TGF-beta signaling, was found to suppress the signaling of both Tranilast and TGF-beta1 to a similar extent. These results suggested that induction of p21 by Tranilast might be closely related to TGF-beta signal transduction pathway.
Collapse
Affiliation(s)
- Min-Ji Charng
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Internal Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chieh-Hsi Wu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Author for correspondence:
| |
Collapse
|
29
|
Wolff RA, Malinowski RL, Heaton NS, Hullett DA, Hoch JR. Transforming growth factor-beta1 antisense treatment of rat vein grafts reduces the accumulation of collagen and increases the accumulation of h-caldesmon. J Vasc Surg 2006; 43:1028-36. [PMID: 16678700 DOI: 10.1016/j.jvs.2006.01.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 01/20/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND The main cause of occlusion and vein graft failure after peripheral and coronary arterial reconstruction is intimal hyperplasia. Transforming growth factor beta-1 (TGF-beta1) is a pleiotropic cytokine known to have powerful effects on cell growth, apoptosis, cell differentiation, and extracellular matrix synthesis. METHODS To investigate the role of TGF-beta1 in intimal hyperplasia, we used adenovirus to deliver to superficial epigastric vein messenger RNA (mRNA) antisense to TGF-beta1 (Ad-AST) or the sequence encoding the bioactive form of TGF-beta1 (Ad-BAT). Infection with "empty" virus was used as a control (Ad-CMVpLpA). The treated vein was then used for an interposition graft into rat femoral artery. Grafts were harvested at 1, 2, 4, and 12 weeks and formalin-fixed for histologic studies or placed in liquid nitrogen for mRNA studies. RESULTS Ad-AST treatment resulted in an overall reduction of TGF-beta1 expression (P = .001), and Ad-BAT treatment resulted in an overall increase in TGF-beta1 expression (P = .007). Histologic analysis showed Ad-AST caused reduced collagen build up in the neointima at 12 weeks (P = .0001). Immunohistochemical staining for h-caldesmon at 12 weeks indicated Ad-AST increased smooth muscle cells throughout the vessel wall compared with Ad-CMVpLpA (P = .0024) or Ad-BAT (P = .04). Ad-AST also resulted in reduced CD68-positive cells in the media/adventitia (P = .005 vs Ad-CMVpLpA, P = .01 vs Ad-BAT). To further understand how Ad-AST was influencing the build up of collagen, we performed quantitative polymerase chain reaction on complimentary DNA (cDNA) from homogenates of the vein grafts. Tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) was increased at 1 week by Ad-BAT (P = .048 vs Ad-CMVpLpA) and decreased by Ad-AST at all time points (P </= .038). The mRNA for collagen-1 alpha-1 was decreased by Ad-AST at 2, 4, and 12 weeks (P < or = .05) and increased by Ad-BAT at 1 week (P = .01). CONCLUSIONS TGF-beta1 antisense treatment of vein grafts prevents the accumulation of collagen in the neointima in part by (1) changing the proportions of the cell types populating the vein graft wall, (2) reducing the mRNA for TIMPs, and (3) reducing the amount of collagen mRNA. With the Ad-AST and Ad-BAT treatments, we have been able to tip the maturation of the vein graft toward positive remodeling (artery-like phenotype) or toward negative remodeling (fibroproliferation and stenosis), respectively.
Collapse
Affiliation(s)
- Randal A Wolff
- William S. Middleton Memorial Veterans Hospital and University of Wisconsin Medical School, Madison, Wisconsin 53792-3236, USA.
| | | | | | | | | |
Collapse
|
30
|
Abstract
Transforming growth factor-beta (TGF-beta) superfamily members, TGF-beta and bone morphogenetic proteins (BMPs), are potent regulatory cytokines with diverse functions on vascular cells. They signal through heteromeric type I and II receptor complexes activating Smad-dependent and Smad-independent signals, which regulate proliferation, differentiation, and survival. They are potent regulators of vascular development and vessel remodeling and play key roles in atherosclerosis and restenosis, regulating endothelial, smooth muscle cell, macrophage, T cell, and probably vascular calcifying cell responses. In atherosclerosis, TGF-beta regulates lesion phenotype by controlling T-cell responses and stimulating smooth muscle cells to produce collagen. It contributes to restenosis by augmenting neointimal cell proliferation and collagen accumulation. Defective TGF-beta signaling in endothelial cells attributable to mutations in endoglin or the type I receptor ALK-1 leads to hereditary hemorrhagic telangiectasia, whereas defective BMP signaling attributable to mutations in the BMP receptor II has been associated with development of primary pulmonary hypertension. The development of mouse models with either cell type-specific or general inactivation of TGF-beta/BMP signaling has started to reveal the importance of the regulatory network of TGF-beta/BMP pathways in vivo and their significance for atherosclerosis, hereditary hemorrhagic telangiectasia, and primary pulmonary hypertension. This review highlights recent findings that have advanced our understanding of the roles of TGF-beta superfamily members in regulating vascular cell responses and provides likely avenues for future research that may lead to novel pharmacological therapies for the treatment or prevention of vascular disorders.
Collapse
Affiliation(s)
- Alex Bobik
- Cell Biology Laboratory, Baker Heart Research Institute, PO Box 6492, St Kilda Rd Central, Melbourne, Victoria 8008, Australia.
| |
Collapse
|
31
|
Qi W, Chen X, Twigg S, Polhill TS, Gilbert RE, Pollock CA. Tranilast attenuates connective tissue growth factor-induced extracellular matrix accumulation in renal cells. Kidney Int 2006; 69:989-95. [PMID: 16528248 DOI: 10.1038/sj.ki.5000189] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tranilast (N-[3,4-dimethoxycinnamoyl]anthranilic acid) is a synthetic compound that we have recently reported to inhibit transforming growth factor-beta1 (TGF-beta1)-induced tubulointerstitial fibrosis in the kidney. Connective tissue growth factor (CTGF) is recognized as a potent downstream mediator of TGF-beta1. Both proximal tubule cells (PTCs) and cortical fibroblasts (CFs) are considered to be responsible for the production of tubulointerstitial extracellular matrix (ECM). These studies were undertaken to assess the profibrotic effects of CTGF in an in vitro model of the human PTCs and CFs, and to determine whether tranilast is effective in limiting the in vitro matrix responses induced by CTGF. Primary cultures of PTCs and CFs were exposed to CTGF (20 ng/ml)+/-tranilast (100 microM). Cell hypertrophy and the secretion of the ECM proteins fibronectin and collagen IV were determined. The effects of tranilast on TGF-beta1-induced CTGF mRNA expression and on phosphorylation of Smad2 were determined. CTGF significantly induced cell hypertrophy, increased fibronectin, and collagen IV secretion in PTCs and CFs. In all cases, the CTGF-induced increase in ECM protein was inhibited in the presence of tranilast. Tranilast reduced CTGF mRNA and phosphorylation of Smad2, which were induced by TGF-beta1 in PTCs and CFs. These results suggest that tranilast is a potential effective antifibrotic compound in the kidney, exerting its effects via inhibition of TGF-beta1-induced CTGF expression and downstream activation of the Smad2 pathway in both PTCs and CFs.
Collapse
Affiliation(s)
- W Qi
- Department of Medicine, Kolling Institute, University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | | | | | | | | | | |
Collapse
|
32
|
Krueger KD, Mitra AK, DelCore MG, Hunter WJ, Agrawal DK. A comparison of stent-induced stenosis in coronary and peripheral arteries. J Clin Pathol 2006; 59:575-9. [PMID: 16473929 PMCID: PMC1860405 DOI: 10.1136/jcp.2004.025643] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND AND OBJECTIVES Restenosis is a complication of interventional procedures such as angioplasty and stenting, often limiting the success of these procedures. Knowledge regarding the relative behaviour of different arteries after these procedures is limited, despite the extensive use of different vascular models. Although the results from studies using different vessels are analysed to predict the behaviour of coronary arteries and other vasculature, direct controlled comparisons between different arteries are necessary for a better understanding of the differential response to restenosis. METHODS This study examines the response to stenting in coronary and internal iliac arteries as characterised by intimal hyperplasia and restenosis. In a swine model of in-stent stenosis, coronary arteries exhibited higher levels of intimal hyperplasia and per cent stenosis than internal iliac arteries. RESULTS After normalisation for injury score, coronary arteries were found to undergo 47% more intimal hyperplasia (p<0.05), whereas per cent stenosis normalised for injury score tended to be higher (p = 0.01). Other measurements reflecting post-stenting intimal hyperplasia (maximal intimal thickness, medial area) did not exhibit significant differences between the artery groups. CONCLUSIONS These results show that coronary vessels are more prone to develop significant intimal hyperplasia and subsequent restenosis than internal iliac vessels. A better insight into how different arteries and arterial components behave is important in understanding and developing newer and better therapeutic measures for restenosis.
Collapse
Affiliation(s)
- K D Krueger
- Department of Medical Microbiology, Creighton University School of Medicine, Omaha, Nebraska 68178, USA
| | | | | | | | | |
Collapse
|
33
|
Abstract
Percutaneous coronary intervention (PCI) has become the most important revascularization method in the treatment of coronary artery disease. The major problem in PCI has been renarrowing of the dilated vessel after the procedure (restenosis). The best results in the prevention of restenosis have been obtained by covering the stent with drugs that inhibit cellular growth, thus limiting excessive scar formation inside of the stent. With drug-eluting stents, restenosis has been reduced to one-tenth compared with balloon angioplasty and to one-fourth compared to bare metal stents. Due to drug-eluting stents, PCI is an alternative to bypass surgery. However, restenosis will remain a challenge due to the increased number of procedures and more difficult disease treated with PCI.
Collapse
Affiliation(s)
- Antti Kivelä
- Department of Medicine, Kuopio University Hospital, Finland
| | | |
Collapse
|
34
|
Zhang QY, Ge JB, Chen JZ, Zhu JH, Zhang LH, Lau CP, Tse HF. Mast cell contributes to cardiomyocyte apoptosis after coronary microembolization. J Histochem Cytochem 2005; 54:515-23. [PMID: 16344327 DOI: 10.1369/jhc.5a6804.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Coronary microembolization (CME) is associated with progressive myocardial dysfunction despite restoration of coronary flow reserve (CFR). The potential pathophysiological role of mast cells (MCs) remains unclear. Therefore, we induced CME in 18 miniswines and determined whether MC accumulation occurs and their effects on local cytokine secretion [interleukin (IL)-6, IL-8, tumor necrosis factor-alpha (TNF-alpha)]; cardiomyocyte apoptosis; and collagen formation at day 1 (D1), day 7 (D7), and day 30 (D30) after CME. Four sham-operated animals without CME (controls) and six animals treated with a MC stabilization agent (tranilast) for 30 days after CME were also studied. CFR decreased at D1 but returned to baseline level at D7 and D30. Coronary sinus levels of IL-6, IL-8, and TNF-alpha increased significantly at D1 and D7 (p<0.01 vs baseline). Levels of IL-6 and IL-8 at D30 returned to baseline level, but not those of TNF-alpha. The numbers of total and degranulating MCs, % apoptotic cardiomyocytes, and collagen volume fraction (CVF) over CME myocardium at D1, D7, and D30 were significantly higher than controls (p<0.01). Treatment with tranilast significantly reduced the serum level of TNF-alpha, numbers of total and degranulating MCs, % apoptotic cardiomyocytes, and CVF at D30 (all p<0.05). There was a significant positive correlation between the numbers of MCs with % apoptotic cardiomyocytes (r = 0.77, p<0.001) and CVF (r = 0.75, p<0.001) over the CME myocardium. Despite restoration of CFR, cardiomyocyte apoptosis persisted after CME and was positively correlated with the number of MCs but was prevented with tranilast treatment. These findings suggest that MCs contribute to cardiomyocyte apoptosis after CME.
Collapse
Affiliation(s)
- Qing-Yong Zhang
- The First Affiliate Hospital of Zhejiang University, HangZhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
35
|
Wolff RA, Ryomoto M, Stark VE, Malinowski R, Tomas JJ, Stinauer MA, Hullett DA, Hoch JR. Antisense to transforming growth factor-β1 messenger RNA reduces vein graft intimal hyperplasia and monocyte chemotactic protein 1. J Vasc Surg 2005; 41:498-508. [PMID: 15838486 DOI: 10.1016/j.jvs.2004.12.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Autogenous vein grafts are commonly used for arterial reconstructive procedures. Their success is limited by the development of intimal hyperplasia (IH), a fibroproliferative disease that predisposes the grafts to occlusive stenosis. Mesenchymal cell proliferation and the deposition of an extracellular matrix characterize neointimal development. Increasing evidence suggests that, regardless of blood vessel type, IH results from complex interactions among vessel wall cells, infiltrating leukocytes, and cytokines. Transforming growth factor-beta1 (TGF-beta1) is a pleiotropic cytokine with powerful effects on inflammatory cell chemotaxis; smooth muscle cell, fibroblast, and endothelial cell proliferation; and extracellular matrix synthesis. METHODS Epigastric vein to common femoral artery interposition grafts were placed in male Lewis rats and harvested at 1, 2, 4, and 12 weeks after surgery. We used replication-defective adenoviruses to deliver a control reporter gene for the enzyme beta-galactosidase (Ad-GAL), empty virus (Ad-CMVpLpA), or the sequence encoding the antisense strand of TGF-beta1 (Ad-AST). The vein graft was transduced passively in medium containing 10 7 plaque-forming units per milliliter of Ad-GAL, Ad-CMVpLpA, or Ad-AST for 20 minutes at room temperature. The adenovirus-treated grafts were compared with grafts treated with medium without virus (sham). RESULTS The Ad-GAL control grafts showed beta-galactosidase activity from 3 days to 4 weeks. Twenty percent of cells were positive out to 2 weeks, at which time the number of cells positive for beta-galactosidase activity began to decline. Treatment with Ad-AST resulted in a significant reduction vs sham, Ad-CMVpLpA, and Ad-GAL in TGF-beta1 messenger RNA, total TGF-beta1 protein, and bioactive TGF-beta1 protein. Neointimal area was significantly reduced in the Ad-AST group vs Ad-GAL at 4 weeks, vs Ad-CMVpLpA at 4 and 12 weeks, and vs sham at 2 and 4 weeks. The medial/adventitial layer was significantly thicker in the Ad-AST group than the Ad-GAL group at 12 weeks. In addition, we studied the effect of Ad-AST on monocyte chemotactic protein 1 (MCP-1). Although the reduction in TGF-beta1 resulted in a reduction of MCP-1 messenger RNA in whole-graft homogenates and MCP-1 protein-positive staining in histologic sections from the perianastomotic region, no reduction in the number of ED1-positive cells (monocytes and macrophages) was observed. CONCLUSIONS Perioperative antisense TGF-beta1 treatment of the vein to be used in arterial reconstructions resulted in a prolonged diminution of IH; this emphasizes the importance of TGF-beta1 in neointimal thickening and indicates that ex vivo gene therapy can reduce the vessel's predisposition to IH. CLINICAL RELEVANCE The main cause of occlusion and graft failure after peripheral and cardiac arterial reconstruction is IH. The study of the mechanisms and mediators of IH, including TGF-beta1, should lead to future gene therapies to prevent or limit IH. The clinical effect of such treatments would be enormous, because they would increase graft longevity, thereby enhancing quality of life and enabling patients to live without the threat of limb loss or recurrent heart attack.
Collapse
Affiliation(s)
- Randal A Wolff
- Department of Surgery, Medical School, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI 53792, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Lee MS, David EM, Makkar RR, Wilentz JR. Molecular and cellular basis of restenosis after percutaneous coronary intervention: the intertwining roles of platelets, leukocytes, and the coagulation-fibrinolysis system. J Pathol 2004; 203:861-70. [PMID: 15258987 DOI: 10.1002/path.1598] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The major limitation of percutaneous coronary intervention (PCI) is restenosis. Restenosis is considered to be an overreaction of the natural healing process after traumatic balloon dilatation. An elaborate web of cellular and molecular responses, including the interaction of platelets, leukocytes, and the coagulation-fibrinolysis system, as well as the secretion of various growth factors and pro-inflammatory cytokines, contributes to neointimal hyperplasia and the development of restenosis. Moreover, platelet and neutrophil activation after stenting appears to be different from that after balloon angioplasty alone. Pharmacotherapy targeting the cell-to-cell interaction between platelets and neutrophils may potentially offer an effective treatment strategy against restenosis after PCI.
Collapse
Affiliation(s)
- Michael S Lee
- Cedars-Sinai Medical Center, UCLA School of Medicine, Cardiovascular Intervention Center, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
37
|
Koch W, Tiroch K, von Beckerath N, Schömig A, Kastrati A. Tumor necrosis factor-alpha, lymphotoxin-alpha, and interleukin-10 gene polymorphisms and restenosis after coronary artery stenting. Cytokine 2004; 24:161-71. [PMID: 14572794 DOI: 10.1016/j.cyto.2003.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammation is the primary response to vessel wall injury caused by stent placement in coronary arteries. The cytokines tumor necrosis factor (TNF)-alpha, lymphotoxin (LT)-alpha, and interleukin (IL)-10 are critically involved in inflammatory reactions. The intensity of the inflammatory process and the angiographic or clinical outcome after stenting are influenced by genetic factors. We investigated the possibility that single nucleotide polymorphisms of the genes encoding TNF-alpha (-863C/A, -308G/A), LT-alpha (252G/A), and IL-10 (-1082G/A, -819C/T, and -592C/A) are associated with the incidence of restenosis, death, or myocardial infarction (MI) after coronary stenting. The gene variations are known to be correlated with transcriptional activity and/or protein production. Our study included 1,850 consecutive patients with symptomatic coronary artery disease who underwent stent implantation. Follow-up angiography was performed in 1,556 patients (84.1%) at six months after the intervention. We found that the polymorphisms are not associated with restenosis, death, or MI. In addition, we did not observe a relationship between polymorphism-specific haplotypes and adverse angiographic and clinical outcomes. In conclusion, functionally relevant polymorphisms of the genes for TNF-alpha, LT-alpha, and IL-10 do not represent genetic markers indicating the risk of restenosis, death, or MI after coronary stenting.
Collapse
Affiliation(s)
- Werner Koch
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstrasse 36, D-80636 Munich, Germany.
| | | | | | | | | |
Collapse
|
38
|
|
39
|
Tardif JC, Grégoire J, Lavoie MA, L'Allier PL. Pharmacologic prevention of both restenosis and atherosclerosis progression: AGI-1067, probucol, statins, folic acid and other therapies. Curr Opin Lipidol 2003; 14:615-20. [PMID: 14624139 DOI: 10.1097/00041433-200312000-00010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW In this article, the authors intend to provide an update on clinical trials of pharmacologic prevention of restenosis after percutaneous coronary interventions, placed in the perspective of the use of orally administered therapy for the prevention of atherosclerosis progression and clinical events. RECENT FINDINGS AGI-1067, the mono-succinic acid ester of probucol, is a phenolic antioxidant member of a novel class of agents termed v-protectants. It has strong antioxidant properties equipotent to those of probucol and antiinflammatory properties. It inhibits gene expression of VCAM-1 and MCP-1 and has been effective at preventing atherosclerosis in all tested animal models including the non-human primate. In the Canadian Antioxidant Restenosis Trial (CART) 1, AGI-1067 and probucol improved lumen dimensions at the site of percutaneous coronary intervention. AGI-1067 also improved luminal dimensions of non-intervened coronary reference segments in the Canadian Antioxidant Restenosis Trial, which suggests a direct antiatherosclerosis effect. Probucol reduced post-percutaneous coronary intervention restenosis and progression of carotid atherosclerosis in other clinical trials. Although statins reduce atherosclerotic events, they do not appear to have a significant effect on restenosis. The failure of folate therapy to protect against restenosis in the Folate After Coronary Intervention Trial (FACIT) occurred despite significant reductions in homocysteine levels. SUMMARY Prevention of both post-percutaneous coronary intervention restenosis and atherosclerosis progression with a pharmacologic agent such as AGI-1067 may be an attractive treatment paradigm. Two important trials that test the antioxidant/antiinflammatory hypothesis are ongoing with AGI-1067: the Canadian Atherosclerosis and Restenosis Trial 2, which assesses its value for the reduction of both atherosclerosis progression and post-percutaneous coronary interventions restenosis, and the Aggressive Reduction of Inflammation Stops Events (ARISE) trial which is evaluating its effects on cardiovascular events.
Collapse
Affiliation(s)
- Jean-Claude Tardif
- Montreal Heart Institute, 5000 Belanger Street, Montreal, PQ, H1T 1C8, Canada.
| | | | | | | |
Collapse
|
40
|
Zhou RH, Lee TS, Tsou TC, Rannou F, Li YS, Chien S, Shyy JYJ. Stent implantation activates Akt in the vessel wall: role of mechanical stretch in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2003; 23:2015-20. [PMID: 12969991 DOI: 10.1161/01.atv.0000095161.06906.ed] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The long-term efficacy of stent implantation is affected by in-stent restenosis (ISR). Multiple factors can contribute to ISR, and the underlying mechanism remains elusive. We investigated the possible role of mechanical stretch and the associated molecular signaling in ISR. METHODS AND RESULTS Stent implantation in rat abdominal aortas induced neointima formation. Immunohistochemical studies revealed the activation of Akt in the media and neointima of the stented vessels. Western blotting showed increased phosphorylation of Akt at both Thr308 and Ser473 and phosphorylation of GSK-3beta in the stented vessels. A stretch device applying static equibiaxial stretch on cultured vascular smooth muscle cells was used to delineate the molecular mechanism underlying the stretch activation of Akt. Static mechanical stretch induced the sustained activation of Akt and its upstream phosphoinositide 3-kinase (PI3K) and the phosphorylation of GSK-3beta, its downstream effector in vascular smooth muscle cells. LY294002, a PI3K inhibitor, and N-acetylcysteine, a scavenger of reactive oxygen species, inhibited the stretch activation of Akt. Furthermore, N-acetylcysteine and wortmannin, another PI3K inhibitor, reduced the neointima formation after stent implantation. CONCLUSIONS Mechanical stretch of the vascular wall during stent deployment may contribute to ISR by activating the Akt pathway.
Collapse
Affiliation(s)
- Rui-Hai Zhou
- Division of Biomedical Sciences, University of California, Riverside, Riverside, Calif 92521-0121, USA
| | | | | | | | | | | | | |
Collapse
|