1
|
Chiu HS, Huang TS, Chen CT, Lin XY, Liao PC, Liou CC, Hsu CC, Somvanshi S, Sumazin P, Hsu PH, Sun CC, Shyu YC. Temporal Regulation of Early-Stage Cytokine Expression in Diabetic Wound Healing Under Negative Pressure Wound Therapy. Int J Mol Sci 2025; 26:4634. [PMID: 40429778 PMCID: PMC12110959 DOI: 10.3390/ijms26104634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/30/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Negative pressure wound therapy (NPWT) is widely recognized for its efficacy in treating diabetic wounds, but the mechanisms involved in the wound healing process remain unclear. By examining changes in blood cytokine levels as molecular signaling precursors, we aim to provide a comprehensive cytokine profile to support adjunctive therapy research and clinical applications. A diabetic mouse wound model was established to compare cytokine profiles between NPWT-treated and standard dressing groups, identifying key signaling candidates that may facilitate wound healing. By integrating normal mouse data with large-scale cytokine analysis, we developed a time-stratified NPWT approach to track acute-phase cytokine fluctuations in diabetic conditions. NPWT did not significantly enhance coagulation-related cytokine expression but effectively reduced inflammation, albeit with a delayed regulatory effect compared to wild-type mice. A one-sided binomial test revealed that NPWT advanced the cytokine expression peak from 16 to 2 h, partially restoring the early healing pattern seen in normal mice and suggesting its potential role in modulating early-stage wound repair. These findings provide novel insights into early cytokine regulation during wound healing and highlight the potential of NPWT to inform therapeutic strategies. This refined monitoring approach may contribute to improved clinical decision-making and support enhanced wound management in diabetic patients.
Collapse
Affiliation(s)
- Hua-Sheng Chiu
- Department of Pediatrics, Texas Children’s Hospital Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (H.-S.C.); (S.S.)
| | - Ting-Shuo Huang
- Department of General Surgery, Jen Ai Hospital, Taichung 400, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Tzung Chen
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- Craniofacial Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Xin-Yu Lin
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan; (X.-Y.L.); (P.-C.L.); (C.-C.L.)
| | - Po-Cheng Liao
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan; (X.-Y.L.); (P.-C.L.); (C.-C.L.)
| | - Cai-Cin Liou
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan; (X.-Y.L.); (P.-C.L.); (C.-C.L.)
| | - Chih-Chin Hsu
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-C.H.); (C.-C.S.)
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
| | - Sonal Somvanshi
- Department of Pediatrics, Texas Children’s Hospital Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (H.-S.C.); (S.S.)
| | - Pavel Sumazin
- Department of Pediatrics, Texas Children’s Hospital Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (H.-S.C.); (S.S.)
| | - Pang-Hung Hsu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Chi-Chin Sun
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-C.H.); (C.-C.S.)
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
| | - Yu-Chiau Shyu
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan; (X.-Y.L.); (P.-C.L.); (C.-C.L.)
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| |
Collapse
|
2
|
Chakraborty R, Borah P, Dutta PP, Sen S. Evolving spectrum of diabetic wound: Mechanistic insights and therapeutic targets. World J Diabetes 2022; 13:696-716. [PMID: 36188143 PMCID: PMC9521443 DOI: 10.4239/wjd.v13.i9.696] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/12/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder resulting in an increased blood glucose level and prolonged hyperglycemia, causes long term health conse-quences. Chronic wound is frequently occurring in diabetes patients due to compromised wound healing capability. Management of wounds in diabetic patients remains a clinical challenge despite many advancements in the field of science and technology. Increasing evidence indicates that alteration of the biochemical milieu resulting from alteration in inflammatory cytokines and matrix metalloproteinase, decrease in fibroblast and keratinocyte functioning, neuropathy, altered leukocyte functioning, infection, etc., plays a significant role in impaired wound healing in diabetic people. Apart from the current pharmacotherapy, different other approaches like the use of conventional drugs, antidiabetic medication, antibiotics, debridement, offloading, platelet-rich plasma, growth factor, oxygen therapy, negative pressure wound therapy, low-level laser, extracorporeal shock wave bioengineered substitute can be considered in the management of diabetic wounds. Drugs/therapeutic strategy that induce angiogenesis and collagen synthesis, inhibition of MMPs, reduction of oxidative stress, controlling hyperglycemia, increase growth factors, regulate inflammatory cytokines, cause NO induction, induce fibroblast and keratinocyte proliferation, control microbial infections are considered important in controlling diabetic wound. Further, medicinal plants and/or phytoconstituents also offer a viable alternative in the treatment of diabetic wound. The focus of the present review is to highlight the molecular and cellular mechanisms, and discuss the drug targets and treatment strategies involved in the diabetic wound.
Collapse
Affiliation(s)
- Raja Chakraborty
- Institute of Pharmacy, Assam Don Bosco University, Kamrup 782402, Assam, India
| | - Pobitra Borah
- School of Pharmacy, Graphic Era Hill University, Dehradun 248002, Uttarakhand, India
| | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam down town University, Guwahati 781026, Assam, India
| | - Saikat Sen
- Faculty of Pharmaceutical Science, Assam down town University, Guwahati 781026, Assam, India
| |
Collapse
|
3
|
Peleli M, Moustakas A, Papapetropoulos A. Endothelial-Tumor Cell Interaction in Brain and CNS Malignancies. Int J Mol Sci 2020; 21:E7371. [PMID: 33036204 PMCID: PMC7582718 DOI: 10.3390/ijms21197371] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma and other brain or CNS malignancies (like neuroblastoma and medulloblastoma) are difficult to treat and are characterized by excessive vascularization that favors further tumor growth. Since the mean overall survival of these types of diseases is low, the finding of new therapeutic approaches is imperative. In this review, we discuss the importance of the interaction between the endothelium and the tumor cells in brain and CNS malignancies. The different mechanisms of formation of new vessels that supply the tumor with nutrients are discussed. We also describe how the tumor cells (TC) alter the endothelial cell (EC) physiology in a way that favors tumorigenesis. In particular, mechanisms of EC-TC interaction are described such as (a) communication using secreted growth factors (i.e., VEGF, TGF-β), (b) intercellular communication through gap junctions (i.e., Cx43), and (c) indirect interaction via intermediate cell types (pericytes, astrocytes, neurons, and immune cells). At the signaling level, we outline the role of important mediators, like the gasotransmitter nitric oxide and different types of reactive oxygen species and the systems producing them. Finally, we briefly discuss the current antiangiogenic therapies used against brain and CNS tumors and the potential of new pharmacological interventions that target the EC-TC interaction.
Collapse
Affiliation(s)
- Maria Peleli
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden;
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden;
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece
| |
Collapse
|
4
|
Karri VVSR, Kuppusamy G, Talluri SV, Yamjala K, Mannemala SS, Malayandi R. Current and emerging therapies in the management of diabetic foot ulcers. Curr Med Res Opin 2016; 32:519-42. [PMID: 26643047 DOI: 10.1185/03007995.2015.1128888] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Diabetic foot ulcers are one of the major causes of mortality in diabetic patients. Very few drugs and therapies have regulatory approval for this indication and several agents from diverse pharmacological classes are currently in various phases of clinical trials for the management of diabetic foot ulcers. SCOPE The purpose of this review is to provide concise information of the drugs and therapies which are approved and present in clinical trials. REVIEW METHODS This review was carried out by systematic searches of relevant guidelines, patents, published articles, reviews and abstracts in PubMed/Medline, Web of Science, clinicaltrials.gov, Cochrane Central Register of Controlled Trials, Cochrane Database of Systematic Reviews and Google Scholar of all English language articles up to 1 March 2015. The following search terms were used: diabetes, diabetic foot, diabetic foot ulcer, diabetic wound, diabetic foot infections, wound management, randomized controlled trials, approved treatments, new treatments and clinical trials. CONCLUSIONS The various drugs and therapies for the management of diabetic foot ulcers comprise antibiotics, neuropathic drugs, wound dressings, skin substitutes, growth factors and inflammatory modulators. The majority of these therapies target the treatment of diabetic foot ulcers to address the altered biochemical composition of the diabetic wound. However, no single treatment can be definitively recommended for the treatment of diabetic foot ulcers.
Collapse
Affiliation(s)
| | - Gowthamarajan Kuppusamy
- a a Department of Pharmaceutics, JSS College of Pharmacy, Ootacamund , JSS University , Mysore , India
| | | | - Karthik Yamjala
- b b Department of Pharmaceutical Analysis, JSS College of Pharmacy, Ootacamund , JSS University , Mysore , India
| | - Sai Sandeep Mannemala
- b b Department of Pharmaceutical Analysis, JSS College of Pharmacy, Ootacamund , JSS University , Mysore , India
- c c Department of Pharmacy, Faculty of Engineering and Technology , Annamalai University , Annamalai Nagar, Tamil Nadu , India
| | - Rajkumar Malayandi
- d d Pharmacokinetic Research and Development, Sun Pharmaceutical Industries Ltd , Baroda , India
- e e JSS College of Pharmacy, Ootacamund , JSS University , Mysore , India
| |
Collapse
|
5
|
The characterization of protein release from sericin film in the presence of an enzyme: Towards fibroblast growth factor-2 delivery. Int J Pharm 2011; 414:193-202. [DOI: 10.1016/j.ijpharm.2011.05.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 04/23/2011] [Accepted: 05/11/2011] [Indexed: 01/02/2023]
|
6
|
Induced interleukin-8 expression in gliomas by tumor-associated macrophages. J Neurooncol 2009; 93:289-301. [PMID: 19156359 DOI: 10.1007/s11060-008-9786-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2008] [Accepted: 12/30/2008] [Indexed: 11/26/2022]
|
7
|
Shieh YS, Hung YJ, Hsieh CB, Chen JS, Chou KC, Liu SY. Tumor-Associated Macrophage Correlated with Angiogenesis and Progression of Mucoepidermoid Carcinoma of Salivary Glands. Ann Surg Oncol 2008; 16:751-60. [DOI: 10.1245/s10434-008-0259-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 11/17/2008] [Accepted: 11/18/2008] [Indexed: 01/17/2023]
|
8
|
Lee CY, Sher HF, Chen HW, Liu CC, Chen CH, Lin CS, Yang PC, Tsay HS, Chen JJW. Anticancer effects of tanshinone I in human non-small cell lung cancer. Mol Cancer Ther 2008; 7:3527-38. [PMID: 19001436 DOI: 10.1158/1535-7163.mct-07-2288] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tanshinones are the major bioactive compounds of Salvia miltiorrhiza Bunge (Danshen) roots, which are used in many therapeutic remedies in Chinese traditional medicine. We investigated the anticancer effects of tanshinones on the highly invasive human lung adenocarcinoma cell line, CL1-5. Tanshinone I significantly inhibited migration, invasion, and gelatinase activity in macrophage-conditioned medium-stimulated CL1-5 cells in vitro and also reduced the tumorigenesis and metastasis in CL1-5-bearing severe combined immunodeficient mice. Unlike tanshinone IIA, which induces cell apoptosis, tanshinone I did not have direct cytotoxicity. Real-time quantitative PCR, luciferase reporter assay, and electrophoretic mobility shift assay revealed that tanshinone I reduces the transcriptional activity of interleukin-8, the angiogenic factor involved in cancer metastasis, by attenuating the DNA-binding activity of activator protein-1 and nuclear factor-kappaB in conditioned medium-stimulated CL1-5 cells. Microarray and pathway analysis of tumor-related genes identified the differentially expressed genes responding to tanshinone I, which may be associated with the Ras-mitogen-activated protein kinase and Rac1 signaling pathways. These results suggest that tanshinone I exhibits anticancer effects both in vitro and in vivo and that these effects are mediated at least partly through the interleukin-8, Ras-mitogen-activated protein kinase, and Rac1 signaling pathways. Although tanshinone I has a remarkable anticancer action, its potential anticoagulant effect should be noted and evaluated.
Collapse
Affiliation(s)
- Chen-Yu Lee
- Institutes of Biomedical Sciences and Molecular Biology, National Chung-Hsing University, Taichung, Taiwan 40227, Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Liu SY, Chang LC, Pan LF, Hung YJ, Lee CH, Shieh YS. Clinicopathologic significance of tumor cell-lined vessel and microenvironment in oral squamous cell carcinoma. Oral Oncol 2008; 44:277-85. [PMID: 17475541 DOI: 10.1016/j.oraloncology.2007.02.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 02/27/2007] [Accepted: 02/27/2007] [Indexed: 11/21/2022]
Abstract
Oral squamous cell carcinoma (SCC) can be vascularized through a process called "tumor cell-lined vessels". Currently, the tumor microenvironment, which is recognized as hypoxic and orchestrated largely by inflammatory cells and defective blood vessels, is considered an important participant in the neoplastic process. We sought to determine their clinicopathologic significance and prognostic implication in oral SCC. Vascular structure was investigated by multistaining with pan-cytokeratin, CD34, and alpha-smooth actin/type IV collagen. Immunohistochemical staining of the hypoxia inducible factor-1 alpha (HIF-1 alpha) and CD68 was used to reflect hypoxia and tumor-associated macrophages (TAM). Our results showed that in a high percentage of vessels in cancer tissue. There is absence of pericyte coverage and loss of basement membrane lining. Significant association between the integrity of vascular structure and lymph node involvement and presence of tumor cell-lined vessel was found. HIF-1 alpha overexpression was frequently observed in cancer cells (78/112) and correlated with tumor progress index. In cancer tissues, the TAM ranged from 28 to 296 cells/mm2 with a mean of 144.6+/-64.3 cells/mm2. There was a significant correlation between TAM and lymph node involvement (P=0.004) and tumor size (P=0.004). Also, a close association was found between TAM count and integrity of vascular structure. In addition, survival analysis revealed that tumor cell-lined vessels (P=0.001), HIF-1 alpha expression (P=0.004), and TAM (P=0.001) correlated significantly with poor survival. We conclude that in the cancer microenvironment, HIF-1 alpha expression and the TAM are induced and contributed to malignant behavior of tumor cells. Furthermore, the presence of tumor cell-lined vessel, HIF-1 alpha overexpression, and high TAM could be the potential markers of prognosis for patients with oral SCC.
Collapse
Affiliation(s)
- Shyun-Yeu Liu
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
10
|
Tanaka E, Ishino Y, Sasaki A, Hasegawa T, Watanabe M, Dalla-Bona DA, Yamano E, van Eijden TMGJ, Tanne K. Fibroblast Growth Factor-2 Augments Recombinant Human Bone Morphogenetic Protein-2-Induced Osteoinductive Activity. Ann Biomed Eng 2006; 34:717-25. [PMID: 16568346 DOI: 10.1007/s10439-006-9092-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2005] [Accepted: 02/16/2006] [Indexed: 10/24/2022]
Abstract
The osteoinductive activity induced by recombinant human BMP-2 (rhBMP-2) blunts proportionately as the recipient ages. In order to compensate for this bluntness administration of fibroblast growth factor-2 (FGF-2) has been considered. The aim of this study was to determine whether FGF-2 administration augments osteoinductive activity caused by rhBMP-2 and to evaluate the effect of aging on bone formation induced by coadministration of rhBMP-2 and FGF-2. Sixty-four Wistar strain male rats of 8-week-old (prepubertal) and 16-week-old (postpubertal) received bone defects bilaterally in the parietal bone and the defects were filled by a polylactic acid polyglycolic acid copolymer/gelatin sponge (PGS) impregnated with rhBMP-2 plus 0 ng, 25 ng, and 250 ng FGF-2 (n=10 in each). At 2 weeks after grafting, the new bone volume seemed to be larger in the rhBMP-2+FGF-2 groups than in the rhBMP-2 alone group. At 4 weeks, the new bone formation was linked to the adjacent original bone. In the prepubertal rats, all newly formed bone was similarly calcified. In the postpubertal rats, only the rhBMP-2+25 ng FGF-2 group showed this higher degree of calcification. At 2 weeks, alkaline phosphatase (ALP) activity in the rhBMP-2+25 ng FGF-2 group was significantly (p<0.05) larger than that in the rhBMP-2 group in both prepubertal and postpubertal rats. This result shows that low-dose administration of FGF-2 enhanced the degree of calcification and ALP activity in the rhBMP-2 grafting site especially in the postpubertal rats. Therefore, FGF-2 would be a candidate to compensate for the reduction of osteoinductive activity of rhBMP-2 with aging.
Collapse
Affiliation(s)
- Eiji Tanaka
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Chen JJW, Lin YC, Yao PL, Yuan A, Chen HY, Shun CT, Tsai MF, Chen CH, Yang PC. Tumor-associated macrophages: the double-edged sword in cancer progression. J Clin Oncol 2004; 23:953-64. [PMID: 15598976 DOI: 10.1200/jco.2005.12.172] [Citation(s) in RCA: 285] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Inflammation plays a critical role in cancer progression. In this study we investigate the pro-tumorigenic activities and gene expression profiles of lung cancer cells after interaction with macrophages. MATERIALS AND METHODS We measured intratumoral microvessel counts and macrophage density in 41 lung cancer tumor specimens and correlated these with the patients' clinical outcome. The interaction between macrophages and cancer cell lines was assessed using a transwell coculture system. The invasive potential was evaluated by in vitro invasion assay. The matrix-degrading activity was assayed by gelatin zymography. The microarray was applied to a large-scale analysis of the genes involved in the interaction, as well as to monitor the gene expression profiles of lung cancer cells responding to anti-inflammatory drugs in cocultures. RESULTS The macrophage density positively correlated with microvessel counts and negatively correlated with patient relapse-free survival (P < .05). After coculture with macrophages, lung cancer cell lines exhibited higher invasive potentials and matrix-degrading activities. We identified 50 genes by microarray that were upregulated more than two-fold in cancer cells after coculture. Northern blot analyses confirmed some gene expression such as interleukin-6, interleukin-8, and matrix metalloproteinase 9. The two-dimensional hierarchical clustering also demonstrated that the gene expression profiles of lung cancer cells responding to various anti-inflammatory drugs in cocultures are distinct. CONCLUSION The interaction of lung cancer cells and macrophages can promote the invasiveness and matrix-degrading activity of cancer cells. Our results also suggest that a great diversity of gene expression occurs in this interaction, which may assist us in understanding the process of cancer metastasis.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Anti-Inflammatory Agents/therapeutic use
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Count
- Cell Line, Tumor
- Coculture Techniques
- Disease Progression
- Disease-Free Survival
- Female
- Gelatinases/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Interleukin-6/genetics
- Interleukin-8/genetics
- Lung Neoplasms/blood supply
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/pathology
- Male
- Matrix Metalloproteinase 9/genetics
- Microcirculation/pathology
- Middle Aged
- Neoplasm Invasiveness
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Sapienza P, di Marzo L, Borrelli V, Sterpetti AV, Mingoli A, Piagnerelli R, Cavallaro A. Basic fibroblast growth factor mediates carotid plaque instability through metalloproteinase-2 and -9 expression. Eur J Vasc Endovasc Surg 2004; 28:89-97. [PMID: 15177237 DOI: 10.1016/j.ejvs.2004.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2004] [Indexed: 10/26/2022]
Abstract
OBJECTIVE(S) We hypothesized that basic fibroblast growth factor (bFGF) may exert a role in carotid plaque instability by regulating the expression of matrix metalloproteinases (MMP). METHODS Plaques obtained from 40 consecutive patients undergoing carotid endarterectomy were preoperatively classified as soft or hard. Serum bFGF was pre- and postoperatively measured. The release of MMP-2 and MMP-9 in the blood serum, and the activity, production and expression in the carotid specimens was analyzed. Specific anti-bFGF inhibition tests were performed in vitro on human umbilical artery smooth muscle cells (HUASMC) to evaluate the role of bFGF in the activity, production and expression of MMP-2 and -9. RESULTS Twenty-one (53%) patients had a soft carotid plaque and 19 (48%) a hard plaque. Preoperative bFGF serum levels were higher in patients with soft plaques [soft=34 (28-39) pg/mL and hard=20 (17-22) pg/mL-p<0.001] and postoperatively returned to normal values (when compared to 10 healthy volunteers). The serum levels of MMP-2 in patients' with soft plaques were higher than those in patients' with hard plaques [soft=1222 (1190-1252) ng/mL and hard=748 (656-793)ng/mL-p<0.0001]. MMP-9 serum values were 26 (22-29) ng/mL for soft plaques and 18 (15-21) ng/mL for hard plaques (p<0.0001). We found increased activity, production and expression of MMP-2 and -9 in soft plaques compared to hard plaques (p<0.001). In vitro inhibition tests on HUASMC showed the direct influence of bFGF on the activity, production and expression of MMP-2 and -9 (p<0.001). CONCLUSIONS bFGF seems to exert a key role in carotid plaque instability regulating the activity, production and expression of MMP thus altering the physiologic homeostasis of the carotid plaque.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/administration & dosage
- Biomarkers/blood
- Blotting, Western
- Carotid Artery, Internal/metabolism
- Carotid Artery, Internal/pathology
- Carotid Stenosis/metabolism
- Dose-Response Relationship, Drug
- Enzyme-Linked Immunosorbent Assay
- Female
- Fibroblast Growth Factor 2/administration & dosage
- Fibroblast Growth Factor 2/metabolism
- Humans
- Immunohistochemistry
- Italy
- Male
- Matrix Metalloproteinase 2/biosynthesis
- Matrix Metalloproteinase 2/drug effects
- Matrix Metalloproteinase 9/biosynthesis
- Matrix Metalloproteinase 9/drug effects
- Matrix Metalloproteinase 9/metabolism
- Middle Aged
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Umbilical Arteries/cytology
- Umbilical Arteries/metabolism
Collapse
Affiliation(s)
- P Sapienza
- Department of Surgery 'Pietro Valdoni', University of Rome 'La Sapienza', Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
13
|
Salcedo R, Oppenheim JJ. Role of chemokines in angiogenesis: CXCL12/SDF-1 and CXCR4 interaction, a key regulator of endothelial cell responses. Microcirculation 2003; 10:359-70. [PMID: 12851652 DOI: 10.1038/sj.mn.7800200] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2002] [Accepted: 02/24/2003] [Indexed: 11/09/2022]
Abstract
Chemokines are small proteins that act as cell attractants via the activation of G protein-coupled receptors. Chemokines play an important role in several pathophysiological processes such as inflammation and immunity. Many proinflammatory chemokines also support the development of vascular blood supply at the site of inflammation. Similarly, tumor-generated chemokines can contribute to tumor growth by promoting angiogenesis. Recently, significant advances have been made in understanding the contribution of chemokines to the angiogenesis process. This review will discuss first the evidence supporting the direct contribution of different chemokine subfamily members, including CC, CXC, and CX3C chemokines, as positive or negative regulators of the angiogenesis process based on the expression of their cognate receptors on endothelial cells. Additionally, the relationship between classic angiogenic factors and chemokine receptor expression on endothelial cells, and the implications of chemokine production by cancer cells will be analyzed with particular emphasis on the CXCL12/stromal-cell derived factor-1 interaction with CXCR4.
Collapse
Affiliation(s)
- Rosalba Salcedo
- Laboratory of Molecular Immunoregulation, Division of Basic Sciences, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | | |
Collapse
|
14
|
Gingras D, Boivin D, Deckers C, Gendron S, Barthomeuf C, Béliveau R. Neovastat--a novel antiangiogenic drug for cancer therapy. Anticancer Drugs 2003; 14:91-6. [PMID: 12569294 DOI: 10.1097/00001813-200302000-00001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neovastat (AE-941) is an antiangiogenic drug isolated from marine cartilage. It interferes with several steps associated with the development of angiogenesis through its ability to induce endothelial cell apoptosis, and to inhibit matrix metalloproteinase activities and vascular endothelial growth factor-mediated signaling pathways, suggesting that Neovastat behaves as a multifunctional antiangiogenic drug. Neovastat is orally bioavailable, and shows significant antitumor and antimetastatic properties in animal models. An excellent safety profile with few side effects has been monitored in more than 800 patients who have been exposed to Neovastat, some of whom for more than 4 years. This indicates that Neovastat is suitable for long-term use, either alone or in combination with other anticancer therapies. Accordingly, Neovastat is currently under evaluation in three pivotal clinical studies with two phase III clinical trials in patients with lung and renal carcinoma, and a phase II clinical trial in patients with multiple myeloma is ongoing.
Collapse
Affiliation(s)
- Denis Gingras
- Laboratoire de médecine moléculaire Hôpital Ste-Justine-UQAM, Centre de cancérologie Charles-Bruneau, Centre de Recherche de l'Hôpital Ste-Justine, Montréal, Québec, Canada
| | | | | | | | | | | |
Collapse
|
15
|
Abstract
It is accepted that novel therapeutic approaches are needed for the majority of patients with malignant brain tumors. The vascularity of many primary brain tumors and the encouraging preclinical studies suggest that antiangiogenic agents have the potential to become an important component of multimodality treatment of patients with brain tumors. The understanding of the biology of angiogenesis is improving rapidly, offering the hope for more specific vascular targeting of brain tumor neovasculature. Neuroimaging techniques evaluating the angiogenic process and the impact of antiangiogenic agents will be an important tool for the rapid development of these novel therapeutic agents.
Collapse
Affiliation(s)
- Michael J Fisher
- Division of Oncology, Children's Hospital of Philadelphia, ARC 907B, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4399, USA
| | | |
Collapse
|
16
|
Fujimura K, Bessho K, Okubo Y, Kusumoto K, Segami N, Iizuka T. The effect of fibroblast growth factor-2 on the osteoinductive activity of recombinant human bone morphogenetic protein-2 in rat muscle. Arch Oral Biol 2002; 47:577-84. [PMID: 12221014 DOI: 10.1016/s0003-9969(02)00046-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
To clarify the effect of recombinant human basic fibroblast growth factor (FGF-2) on the osteoinductive activity of recombinant human bone morphogenetic protein-2 (BMP-2) in vivo, different amounts of FGF-2 (0, 16, 80 and 400 ng, and 2, 10 and 50 micro g: n=10 in each group), BMP-2 (2 micro g) and type I collagen as a carrier were mixed and implanted into rat calf muscles. Three weeks after implantation, compared with the controls, the radiopaque shadows of the implants were increased in the 16, 80 and 400 ng FGF-2-treated groups, but decreased in the 2, 10 and 50 micro g FGF-2-treated groups. In addition, alkaline phosphatase activity was increased in the 16, 80 and 400 ng FGF-2-treated groups but decreased in the 50 micro g FGF-2-treated group. Histological examination revealed increased bone formation in the 16, 80 and 400 ng FGF-2-treated groups. These results show that combined treatment with FGF-2 and BMP-2 has a biphasic effect on osteoinductive activity, i.e. it increases with low doses of FGF-2 and decreases with high doses of FGF-2.
Collapse
Affiliation(s)
- Kazuma Fujimura
- Department of Oral and Maxillofacial Surgery, Kanazawa Medical University, Ishikawa Pref 920-0293, Japan.
| | | | | | | | | | | |
Collapse
|
17
|
Gingras D, Batist G, Béliveau R. AE-941 (Neovastat): a novel multifunctional antiangiogenic compound. Expert Rev Anticancer Ther 2001; 1:341-7. [PMID: 12113101 DOI: 10.1586/14737140.1.3.341] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
AE-941 (Neovastat) is a naturally occurring product extracted from cartilage and has antiangiogenic properties. It has reached Phase III clinical trial evaluation for the treatment of solid tumors (non-small cell lung cancer and renal cell carcinoma) and a pivotal Phase II clinical trial in multiple myeloma is ongoing. AE-941 inhibits several steps of the angiogenesis process, including matrix metalloproteinase activities and VEGF signaling pathways. Moreover, AE-941 induces endothelial cell apoptosis and tissue-type plasminogen activator activity, thus suggesting that it is a multifunctional antiangiogenic drug. Results from Phase I/II clinical trials indicate that AE-941, given orally, is well tolerated. Moreover, the median survival time in patients with renal cell carcinoma and non-small cell lung cancer was significantly longer in patients receiving high doses of AE-941 compared to low doses.
Collapse
Affiliation(s)
- D Gingras
- Laboratiore de médicine moléculaire Hôpital Ste-Justine-UQAM, Centre de cancérologie Charles-Bruneau, Centre de Recherche de l'Hôpital Ste-Justine, 3175, Chemin Côte-Ste-Catherine, Montréal, Québec H3T 1C5, Canada
| | | | | |
Collapse
|
18
|
Crowther M, Brown NJ, Bishop ET, Lewis CE. Microenvironmental influence on macrophage regulation of angiogenesis in wounds and malignant tumors. J Leukoc Biol 2001; 70:478-490. [PMID: 11590184 DOI: 10.1189/jlb.70.4.478] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Angiogenesis is the development of blood vessels from an existing vasculature. This process is fundamental to both physiological wound healing and the growth of malignant tumors, as it restores or creates a blood supply to growing tissue. In both cases, the release of angiogenic molecules by macrophages recruited to the wound or tumor site is central to the formation of these neovessels. Reduced vascular perfusion in tissues generates tissue ischemia and a marked reduction in local levels of oxygen (hypoxia) and glucose. Cells adapt by switching to anaerobic metabolic pathways, with a concomitant increase in lactate production and reduction in extracellular pH. In tumors, these microenvironmental "stress" factors stimulate tumor cells to secrete a wide array of proangiogenic cytokines and enzymes, promoting the re-establishment of a local vascular supply. Here we review the evidence that these stress factors, in particular hypoxia and high lactate levels, stimulate macrophages to perform similar proangiogenic functions in both tumors and wounds. The resolution of wounds results in restoration of tissue integrity and perfusion, and macrophage presence is reduced to preinjury levels. However, in tumors a high number of macrophages persists and might contribute to the ongoing growth, neovascularization, and metastasis of malignant cells.
Collapse
Affiliation(s)
- M Crowther
- Tumor Targeting Group, Section of Oncology & Pathology, Division of Genomic Medicine, University of Sheffield Medical School, Sheffield S10 2RX, United Kingdom
| | | | | | | |
Collapse
|
19
|
Abstract
It has become well accepted that solid tumors must create a vascular system for nutrient delivery and waste removal in order to grow appreciably. This process, angiogenesis, is critical to the progression of gliomas, with vascular changes accompanying the advancement of these tumors. The cascade of events in this process of blood vessel formation involves a complex interplay between tumor cells, endothelial cells, and their surrounding basement membranes in which enzymatic degradation of surrounding ground substance and subsequent endothelial cell migration, proliferation, and tube formation occurs. It is likely that a host of growth factors is responsible for mediating these key events. To date, a role for Vascular Endothelial Growth Factor (VEGF) in glioma angiogenesis has been convincingly demonstrated. This review explores the contribution of other growth factors--Fibroblast Growth Factors (FGFs), Platelet-Derived Growth Factor (PDGF), Epidermal Growth Factor (EGF), and Transforming Growth Factors (TGFs)--to glioma angiogenesis. These growth factors may influence glioma angiogenesis by directly stimulating endothelial cell proliferation, by mediating the expression of key proteases on endothelial cells necessary for angiogenesis, or by regulating the expression of VEGF and of each other.
Collapse
Affiliation(s)
- I F Dunn
- Brain Tumor Research Center, Brigham and Women's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
20
|
Mackarel AJ, Cottell DC, Fitzgerald MX, O'Connor CM. Human endothelial cells cultured on microporous filters used for leukocyte transmigration studies form monolayers on both sides of the filter. In Vitro Cell Dev Biol Anim 1999; 35:346-51. [PMID: 10476922 DOI: 10.1007/s11626-999-0085-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A growing number of studies on the mechanism of leukocyte transendothelial migration use endothelial cells grown on microporous filters as an in vitro model of endothelium. Ultrastructural examination of such a model system previously demonstrated that human pulmonary artery endothelial cells (HPAEC) formed confluent monolayers on both sides of the 3-microm-pore filter (Mackarel et al., 1999). To determine whether this was a characteristic specific to pulmonary artery endothelial cells, the growth characteristics of a human pulmonary microvascular endothelial cell type (HMVEC-L) and the widely used human umbilical vein endothelial cells (HUVEC) on 3-microm microporous filters were examined by transmission electron microscopy (TEM). Similar to HPAEC, HMVEC-L and HUVEC were also found to grow on both sides of the filter. All three endothelial cell types were capable of migrating through the 3 microm pores of the filter to form a monolayer on the filter underside. The endothelial cells on the underside were orientated in an inverted position with the luminal surface facing away from the filter. Such 'bilayer' formation was observed at a range of seeding densities and in different culture media. Despite the presence of a bilayer of endothelial cells, TEM demonstrated that neutrophils migrated successfully across the cell-filter-cell system. Previous transmigration reports in which an in vitro model similar to ours was used have often assumed only one layer of endothelial cells. The observations reported here indicate that while endothelial cells on microporous filters are useful models for examining leukocyte-endothelial interactions, they are not appropriate for studies examining endothelial cell 'sidedness.'
Collapse
Affiliation(s)
- A J Mackarel
- Department of Medicine and Therapeutics, University College Dublin, Ireland
| | | | | | | |
Collapse
|
21
|
Cucina A, Corvino V, Sapienza P, Borrelli V, Lucarelli M, Scarpa S, Strom R, Santoro-D'Angelo L, Cavallaro A. Nicotine regulates basic fibroblastic growth factor and transforming growth factor beta1 production in endothelial cells. Biochem Biophys Res Commun 1999; 257:306-12. [PMID: 10198208 DOI: 10.1006/bbrc.1999.0478] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicotine, a constituent of cigarette smoking, may induce atherosclerosis through the production of growth factors. The pattern of bFGF and TGF beta1 production and release by bovine aortic endothelial cells (EC) stimulated with nicotine (from 6 x 10(-4) to 6 x 10(-8) M) was studied. EC viability and count were assessed. The presence of bFGF and TGF beta1 in serum-free conditioned media was determined by the inhibition antibody-binding assay and Western blot analysis. Mitogenic activity of nicotine on EC was also determined. Polymerase chain reaction (PCR) was used to study the expression of bFGF and TGF beta1. The bFGF release after nicotine stimulation was greater than controls, whereas TGF beta1 release was lower. At a nicotine concentration of 6 x 10(-6) M we noted the greatest mitogenic activity. The addition of monoclonal antibody anti-bFGF decreased the tritiated thymidine uptake of EC exposed to nicotine but the addition of monoclonal antibody anti-TGF beta1 had no significant effect. bFGF mRNA expression was significantly higher in EC exposed to nicotine than in controls, whereas TGF beta1 mRNA expression was not modified. From these data we concluded that nicotine regulates bFGF production and release and TGF beta1 release and may have a key role in the development and progression of atherosclerosis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Aorta
- Arteriosclerosis/etiology
- Arteriosclerosis/metabolism
- Blotting, Western
- Cattle
- Cell Count/drug effects
- Cell Division/drug effects
- Cell Survival/drug effects
- Culture Media, Conditioned/pharmacology
- DNA/biosynthesis
- Dose-Response Relationship, Drug
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/immunology
- Fibroblast Growth Factor 2/metabolism
- Mitogens/metabolism
- Mitogens/pharmacology
- Muscle, Smooth/cytology
- Muscle, Smooth/drug effects
- Nicotine/pharmacology
- Nicotine/toxicity
- Polymerase Chain Reaction
- RNA, Messenger/metabolism
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- A Cucina
- 1st Department of Surgery, University of Rome "La Sapienza,", Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Klominek J, Baskin B, Liu Z, Hauzenberger D. Hepatocyte growth factor/scatter factor stimulates chemotaxis and growth of malignant mesothelioma cells through c-met receptor. Int J Cancer 1998; 76:240-9. [PMID: 9537587 DOI: 10.1002/(sici)1097-0215(19980413)76:2<240::aid-ijc12>3.0.co;2-g] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Hepatocyte growth factor (HGF) and its receptor c-met are present in several human tissues but their expression in mesothelial cells has not been examined. In this study, we have investigated the expression of HGF and c-met in normal human mesothelial cells and 11 human malignant mesothelioma cell lines. Using RT-PCR and Western blotting we found that HGF is produced by 3/11 mesothelioma cell lines whereas c-met is expressed in 11/11 mesothelioma cell lines. In addition, c-met expression was also found in 6/6 cell samples obtained from pleural fluids of patients with mesothelioma. In contrast, neither normal cultured mesothelial cells nor mesothelial cells obtained directly from patients without mesothelioma expressed HGF nor c-met. We have also analysed the biological function of HGF and c-met in mesothelioma cell lines. Recombinant human (rh) HGF stimulated both directional (chemotactic) and random (chemokinetic) motility in all mesothelioma cell lines tested. Furthermore, mesothelioma serum free conditioned medium containing HGF stimulated mesothelioma cell migration. This effect could be blocked in the presence of neutralizing anti-HGF monoclonal antibodies (MAbs) in the assay. Addition of HGF to mesothelioma cells cultured on collagen type IV was associated with induction of bipolar shape and protrusion of prominent pseudopodia. We have also found that rhHGF was mitogenic for mesothelioma cells. Our findings suggest that expression of HGF/c-met is involved not only in mesothelioma progression but also in its growth and migration and that c-met expression found in mesothelioma cells taken directly from patients may be of diagnostic importance.
Collapse
Affiliation(s)
- J Klominek
- Department of Lung Medicine, Karolinska Institute at Huddinge Hospital, Sweden.
| | | | | | | |
Collapse
|
23
|
Plyley MJ, Olmstead BJ, Noble EG. Time course of changes in capillarization in hypertrophied rat plantaris muscle. J Appl Physiol (1985) 1998; 84:902-7. [PMID: 9480950 DOI: 10.1152/jappl.1998.84.3.902] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The time course of angiogenesis during hypertrophy of the rat plantaris muscle was studied by using a unilateral, synergistic ablation model. Animals (n = 6/group) were euthanized 2, 5, 7, 15, 21, and 30 days postmyectomy. Sections from both the hypertrophied and contralateral muscles were simultaneously stained for capillaries and muscle fiber type. Mean fiber cross-sectional area (FA) and various indexes of capillarity were determined by using a video analysis system. The capillary supply to individual fibers, assessed as the FA supplied per capillary contact, remained unchanged until day 21 (compared with day 2) and exhibited a significant increase at day 30. Analysis of the time course of capillary development on the basis of the number of capillary contacts per fiber, and of hypertrophy on the basis of FA, yielded half-lives of 10.1 and 11.2 days, respectively. It was concluded that angiogenesis during muscle overload is tightly coupled to the changes in FA, which could suggest that the two processes are initiated and/or driven by some common factor(s).
Collapse
Affiliation(s)
- M J Plyley
- Faculty of Kinesiology, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | | | | |
Collapse
|
24
|
Gade PV, Andrades JA, Nimni ME, Becerra J, Longoria J, Asemanfar N, Sorgente N. Nitric oxide mediates hyperglycemia-induced defective migration in cultured endothelial cells. J Vasc Surg 1997; 26:319-26. [PMID: 9279321 DOI: 10.1016/s0741-5214(97)70195-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE To examine the effects of elevated glucose on the migration and proliferation of vascular endothelial cells in an in vitro wound model and to investigate whether nitric oxide (NO) mediates the effects of elevated glucose. METHODS Migration was investigated in monolayers of bovine aortic endothelial cells wounded by scraping and measuring the distance, the number of cells migrating, and the area covered by the migrating cells in the presence of various concentrations of glucose. The effects of NO were evaluated by adding to the cultures NG-monomethyl arginine (NMMA), an inhibitor of NO synthase, or S-nitrosylated penicillamine, which is a slow-release agent of NO. Proliferation was investigated in the presence of various concentrations of serum, glucose, or both. RESULTS Elevated glucose levels (16.5 and 27.7 mmol/L) inhibited endothelial cell migration in a dose-dependent manner compared with cells cultured in the presence of 5.5 mmol/L glucose. Inhibition of migration was also observed when wounded mono-layers cultured in 5.5 mmol/L glucose were treated with S-nitrosylated penicillamine, which generates NO. Inhibition of NO synthase by NMMA prevented the inhibition of migration observed in media containing 27.7 mmol/L glucose. Elevated glucose levels did not affect cell proliferation except in the presence of 20% fetal bovine serum. CONCLUSIONS An elevated glucose level inhibits endothelial cell migration in an in vitro wound model, and the inhibition appears to be mediated by increased levels of NO.
Collapse
Affiliation(s)
- P V Gade
- Department of Surgery, Maimonides Medical Center, Brooklyn, N.Y., USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Kuo NT, Norman JT, Wilson PD. Acidic FGF regulation of hyperproliferation of fibroblasts in human autosomal dominant polycystic kidney disease. BIOCHEMICAL AND MOLECULAR MEDICINE 1997; 61:178-91. [PMID: 9259983 DOI: 10.1006/bmme.1997.2583] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by cystic tubule enlargement and expansion of the interstitium associated with fibrosis. Our previous studies have analyzed the increased proliferation of cystic epithelial cells and this study examines the basis of increased proliferation of interstitial fibroblasts associated with ADPKD disease progression. ADPKD fibroblasts show phenotypic alterations in vitro, have acquired the capacity to grow in soft agar, and show an increased mitogenic response to a variety of growth factors particularly acidic FGF (aFGF). ELISA, Western immunoblot analysis, and immunocytochemistry showed increased aFGF content in ADPKD tissues and fibroblasts in culture, and aFGF was secreted into the extracellular matrix and conditioned medium, respectively. No alterations in aFGF receptor number were found, but Scatchard analysis of 125I-aFGF binding suggested an increased affinity of binding to the low affinity receptor, and covalent cross-linking analysis suggested the presence of novel putative receptors (120 kDa) in ADPKD fibroblasts. Signaling abnormalities were found, since aFGF incubation resulted in the tyrosine phosphorylation of additional substrates, more rapidly and for a more sustained duration in ADPKD fibroblasts than in normal fibroblasts. These findings suggest an important role for acidic FGF in the hyperproliferation of interstitial fibroblasts associated with disease progression in human ADPKD.
Collapse
Affiliation(s)
- N T Kuo
- Department of Physiology, UMDNJ-RWJ (formerly Rutgers) Medical School, Piscataway 08854, USA
| | | | | |
Collapse
|
26
|
Kähler CM, Kirchmair R, Kaufmann G, Kähler ST, Reinisch N, Fischer-Colbrie R, Hogue-Angeletti R, Winkler H, Wiedermann CJ. Inhibition of proliferation and stimulation of migration of endothelial cells by secretoneurin in vitro. Arterioscler Thromb Vasc Biol 1997; 17:932-9. [PMID: 9157958 DOI: 10.1161/01.atv.17.5.932] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Vascular cell responses in inflammation are affected by several neuropeptides of perivascular nerve fibers. Secretoneurin is a 33-amino acid peptide that is coreleased from these nerve endings with other proinflammatory neuropeptides, eg, substance P and calcitonin gene-related peptide. Furthermore, secretoneurin has been shown to be chemotactic for human skin fibroblasts and human blood monocytes in vitro and in vivo. An action on cellular components of the vascular wall is not yet reported. We therefore investigated in vitro effects of this novel sensory neuropeptide on endothelial cells. Secretoneurin exerted a potent and reversible inhibitory effect both on endothelial cell growth under low serum conditions (1% fetal calf serum) and endothelial cell growth factor-activated endothelial cell proliferation. We show in the present study that secretoneurin exerts this effect on aortic (rat) and pulmonary artery (bovine) endothelial cells, as well as venous (human umbilical vein) endothelium. Endothelial cell chemotaxis was tested by means of three different migration assays employing nitrocellulose and polycarbonate micropore filters. Secretoneurin consistently exhibited potent chemoattractant activity. The effective concentrations for the observed effects were in the picomolar range. The combination of chemotactic and antiproliferative effects on endothelial cells suggests that secretoneurin may act as a regulatory factor of vascular cell functions.
Collapse
Affiliation(s)
- C M Kähler
- Department of Internal Medicine, Faculty of Medicine, University of Innsbruck, Anichstrasse, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sigounas G, Steiner M, Anagnostou A. Synergism of hemopoietic growth factors on endothelial cell proliferation. Angiology 1997; 48:141-7. [PMID: 9040269 DOI: 10.1177/000331979704800207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Endothelial cells, which are nonhemopoietic cells, express and/or produce most of the known hemopoietic receptors and cytokines. The biological role of these factors, and their respective receptors, on endothelial cells is still unknown. In this study, the authors assessed the effect of different hemopoietic growth factors, ie, interleukin-3 (IL-3), erythropoietin (EPO), macrophage-colony stimulating factor (M-CSF), granulocyte-colony stimulating factor (G-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), singly or in conjunction with others, on proliferation and chemotaxis of human umbilical vein endothelial cells (HUVECs). They found growth stimulatory activity with IL-3, EPO, and GM-CSF and potent synergism between EPO and IL-3, less with IL-3 and GM-CSF, and none with EPO and either GM-CSF or G-CSF. All the singly tested hemopoietic growth factors stimulated the migration of HUVECs, but in conjunction with other factors, they did not show any additive or synergistic effect.
Collapse
Affiliation(s)
- G Sigounas
- Department of Medicine, East Carolina University School of Medicine, Greenville, North Carolina, USA
| | | | | |
Collapse
|
28
|
Klein S, Roghani M, Rifkin DB. Fibroblast growth factors as angiogenesis factors: new insights into their mechanism of action. EXS 1997; 79:159-92. [PMID: 9002232 DOI: 10.1007/978-3-0348-9006-9_7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- S Klein
- Department of Cell Biology, New York University Medical Center, New York 10016, USA
| | | | | |
Collapse
|
29
|
Abstract
We have examined the binding of FGF-2 to ribosomes and have found that in NIH 3T3 cells that synthesize high amounts of all FGF-2 forms, both 18 kDa and HMW forms of FGF-2 bind to ribosomes. Ribosomes purified from these cells were treated with RNase or puromycin to identify the binding site of FGF-2 on the ribosome. Neither RNase nor puromycin treatment affected the in vivo binding of FGF-2 to ribosomes suggesting that FGF-2 binds ribosomal protein or rRNA, but not mRNA. The stoichiometry of binding in these cells was approximately 1 FGF-2 molecule bound per 1 ribosome. Binding was unaffected by high salt treatment indicating that FGF-2 tightly associates with polysomes. An in vitro binding experiment performed with purified ribosomes and recombinant FGF-2 suggested that the binding site is saturable. HBNF, a protein with similar charge and size to FGF-2, bound 15-fold less than FGF-2 to purified ribosomes. These results indicate that the binding of FGF-2 to ribosomes is specific.
Collapse
Affiliation(s)
- S Klein
- Department of Cell Biology, Raymond and Beverly Sackler Foundation Laboratory, New York University Medical Center, New York, NY 10016, USA
| | | | | |
Collapse
|
30
|
Bilato C, Pauly RR, Melillo G, Monticone R, Gorelick-Feldman D, Gluzband YA, Sollott SJ, Ziman B, Lakatta EG, Crow MT. Intracellular signaling pathways required for rat vascular smooth muscle cell migration. Interactions between basic fibroblast growth factor and platelet-derived growth factor. J Clin Invest 1995; 96:1905-15. [PMID: 7560082 PMCID: PMC185827 DOI: 10.1172/jci118236] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Intracellular signaling pathways activated by both PDGF and basic fibroblast growth factor (bFGF) have been implicated in the migration of vascular smooth muscle cells (VSMC), a key step in the pathogenesis of many vascular diseases. We demonstrate here that, while bFGF is a weak chemoattractant for VSMCs, it is required for the PDGF-directed migration of VSMCs and the activation of calcium/calmodulin-dependent protein kinase II (CamKinase II), an intracellular event that we have previously shown to be important in the regulation of VSMC migration. Neutralizing antibodies to bFGF caused a dramatic reduction in the size of the intracellular calcium transient normally seen after PDGF stimulation and inhibited both PDGF-directed VSMC migration and CamKinase II activation. Partially restoring the calcium transient with ionomycin restored migration and CamKinase II activation as did the forced expression of a mutant CamKinase II that had been "locked" in the active state by site-directed mutagenesis. These results suggest that bFGF links PDGF receptor stimulation to changes in intracellular calcium and CamKinase II activation, reinforcing the central role played by CamKinase II in regulating VSMC migration.
Collapse
Affiliation(s)
- C Bilato
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Futami K, Yamashita J, Tachibana O, Kida S, Higashi S, Ikeda K, Yamashima T. Basic fibroblast growth factor may repair experimental cerebral aneurysms in rats. Stroke 1995; 26:1649-54. [PMID: 7660412 DOI: 10.1161/01.str.26.9.1649] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE To determine whether basic fibroblast growth factor (FGF) can induce proliferative response of endothelial cells and/or smooth muscle cells in aneurysmal lesions, we investigated the effect of the intravenous administration of basic FGF on experimental cerebral aneurysms. METHODS Cerebral aneurysms were induced in rats by ligation of the unilateral common carotid artery, producing hypertension. Three months later, basic FGF was intravenously injected in two groups of randomly divided rats on days 1, 3, and 5 at two different doses (low dose: 2 micrograms/100 g body wt per day; high dose: 5 micrograms/100 g body wt per day). In a control group, normal saline was similarly injected. The junctions of the anterior cerebral artery (ACA) and the olfactory artery (OA) were examined with a light microscope. Aneurysmal changes were defined as the lesions with discontinuity of the internal elastic lamina in more than half of the outward dilated wall. Depending on whether the smooth muscle cell layer was present in the whole wall, the lesions were divided into two stages: early aneurysmal lesion (whole area) and saccular aneurysm (not totally preserved). RESULTS The control and the low-dose groups presented no obvious intimal thickening in the intact ACA-OA junctions of both nonligated and ligated sides as well as in the aneurysmal changes. In contrast, in the high-dose group, various degrees of intimal thickening in the wall were detected in 7 of 15 early aneurysmal lesions (P = .019, Fisher's exact test). Immunohistochemistry showed the proliferated cells to be smooth muscle cells. CONCLUSIONS These results demonstrate that exogenous basic FGF induces the proliferative response of smooth muscle cells in aneurysmal lesions in rats.
Collapse
Affiliation(s)
- K Futami
- Department of Neurosurgery, Kanazawa University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Harada K, Grossman W, Friedman M, Edelman ER, Prasad PV, Keighley CS, Manning WJ, Sellke FW, Simons M. Basic fibroblast growth factor improves myocardial function in chronically ischemic porcine hearts. J Clin Invest 1994; 94:623-30. [PMID: 7518840 PMCID: PMC296139 DOI: 10.1172/jci117378] [Citation(s) in RCA: 217] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The effect of basic fibroblast growth factor (bFGF) administration on regional myocardial function and blood flow in chronically ischemic hearts was studied in 26 pigs instrumented with proximal circumflex coronary artery (LCX) ameroid constrictors. In 13 animals bFGF was administered extraluminally to the proximal left anterior descending (LAD) and LCX arteries with heparin-alginate beads and 13 other animal served as controls. bFGF-treated pigs showed a fourfold reduction in left ventricular infarct size compared to untreated controls (infarct size: 1.2 +/- 0.4% vs. 5.1 +/- 1.3% of LV mass, mean +/- SEM, P < 0.05). Percent fractional shortening (% FS) in the LCX area at rest was reduced compared with the LAD region in both bFGF and control pigs. However, there was better recovery in the LCX area after rapid pacing in bFGF-treated pigs (% FSLCX/% FSLAD, 22.9 +/- 7.3%-->30.5 +/- 8.5%, P < 0.05 vs. prepacing) than in controls (16.0 +/- 7.8%-->14.3 +/- 7.0%, P = NS). Furthermore, LV end-diastolic pressure rise with rapid pacing was less in bFGF-treated than control pigs (pre-pacing; pacing; post-pacing, 10 +/- 1; 17 +/- 3; 11 +/- 1* mmHg vs 10 +/- 1; 24 +/- 4; 15 +/- 1 mmHg, *P < 0.05 vs. control). Coronary blood flow in the LCX territory (normalized for LAD flow) was also better during pacing in bFGF-treated pigs than in controls. Thus, periadventitial administration of bFGF in a gradual coronary occlusion model in pigs results in improvement of coronary flow and reduction in infarct size in the compromised territory as well as in prevention of pacing-induced hemodynamic deterioration.
Collapse
Affiliation(s)
- K Harada
- Charles A. Dana Research Institute, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Jampol LM, Ebroon DA, Goldbaum MH. Peripheral proliferative retinopathies: an update on angiogenesis, etiologies and management. Surv Ophthalmol 1994; 38:519-40. [PMID: 8066542 DOI: 10.1016/0039-6257(94)90146-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Many clinical entities may be associated with the development of peripheral retinal neovascularization. In this paper, we review the mechanisms of normal and abnormal angiogenesis in the retina. Specific disease entities associated with peripheral proliferative retinopathies are discussed. These include vascular disease with ischemia, inflammatory diseases with possible ischemia and a variety of miscellaneous causes, including hereditary diseases and tumors. Basic principles for the clinical evaluation of patients with retinal neovascularization are described. Finally, the treatments for retinal neovascularization, including cryopexy and local and panretinal photocoagulation are reviewed, and techniques and possible mechanisms of the beneficial results of treatment are described.
Collapse
Affiliation(s)
- L M Jampol
- Department of Ophthalmology, Northwestern University Medical School, Chicago, Illinois
| | | | | |
Collapse
|
34
|
Biro S, Yu ZX, Fu YM, Smale G, Sasse J, Sanchez J, Ferrans VJ, Casscells W. Expression and subcellular distribution of basic fibroblast growth factor are regulated during migration of endothelial cells. Circ Res 1994; 74:485-94. [PMID: 8118957 DOI: 10.1161/01.res.74.3.485] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Migration of endothelial cells is involved in normal and pathological angiogenesis and in re-endothelialization after vascular injury or rupture of atherosclerotic plaques. Several types of endothelial cells are known to synthesize basic fibroblast growth factor (bFGF); in some of these, migration is increased by exogenous bFGF and inhibited by anti-bFGF antibodies. Using immunocytochemical techniques and RNase protection analysis, we studied endothelial cells from bovine coronary arteries and veins as well as from adrenal microvessels. We found that bFGF mRNA and peptide were present in confluent endothelial cells and were upregulated during migration stimulated by removal of some cells from the monolayer. During migration, extracellular matrix stores of bFGF were depleted, and bFGF immunoreactivity began to accumulate in the cytoplasm of endothelial cells between 2 and 6 hours. After migration had begun, but before the initiation of DNA synthesis, bFGF immunoreactivity increased in the nuclei and nucleoli. Exogenous bFGF stimulated endothelial migration, and antibodies to bFGF markedly inhibited migration, suggesting that an intracrine function of nuclear bFGF is not sufficient for cell migration. In all three types of endothelial cells studied, bFGF was identified as an endogenous regulator, but not as the sole regulator, or migration. Moreover, bFGF expression and subcellular localization were found to be regulated during endothelial cell migration.
Collapse
Affiliation(s)
- S Biro
- Cardiology Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Nugent MA, Karnovsky MJ, Edelman ER. Vascular cell-derived heparan sulfate shows coupled inhibition of basic fibroblast growth factor binding and mitogenesis in vascular smooth muscle cells. Circ Res 1993; 73:1051-60. [PMID: 8222077 DOI: 10.1161/01.res.73.6.1051] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Basic fibroblast growth factor (bFGF) has been previously shown to be mitogenic for vascular smooth muscle cells (VSMCs) in vivo, but only after vascular injury. We show in the present study that the regulation of bFGF-stimulated VSMC proliferation, by vascular cell-secreted heparin-like compounds, correlates with inhibition of bFGF binding to cell-associated heparin sulfate proteoglycans. The stimulation of cultured VSMC proliferation by bFGF was markedly reduced when these cells were cocultured with confluent endothelial cells or confluent VSMCs (100.8 +/- 8.4% and 55.6 +/- 2.3% inhibition, respectively) or with conditioned media from these two cell types. Balb/c3T3 fibroblasts had no statistically significant effect on bFGF-stimulated VSMC proliferation. Vascular cell-conditioned media also inhibited bFGF binding to heparan sulfate proteoglycans on VSMCs, and the inhibition of binding correlated linearly with the inhibition of proliferation after a critical amount of binding was inhibited (44%) (r = .952, P < .0001). Heparinase or heparitinase treatment of conditioned media removed the bFGF-inhibitory effects, presumably by degrading heparin-like compounds. Indeed, heparin itself mimicked the inhibitory effects of conditioned media on bFGF-mediated proliferation and binding to heparin sulfate proteoglycans. These results suggest a bFGF regulatory role for vascular cell-produced heparin-like compounds, linking the mitogenic effects with binding to heparan sulfate proteoglycans for this heparin-binding growth factor.
Collapse
Affiliation(s)
- M A Nugent
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge
| | | | | |
Collapse
|
36
|
Edelman ER, Nugent MA, Karnovsky MJ. Perivascular and intravenous administration of basic fibroblast growth factor: vascular and solid organ deposition. Proc Natl Acad Sci U S A 1993; 90:1513-7. [PMID: 8434012 PMCID: PMC45904 DOI: 10.1073/pnas.90.4.1513] [Citation(s) in RCA: 192] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The in vivo mitogenicity of basic fibroblast growth factor (bFGF) for arterial smooth muscle cells relies on the removal of endothelium, raising the question of whether the endothelium serves as a mechanical barrier preventing contact of circulating bFGF with underlying smooth muscle cells or as a biochemical barrier that produces a local inhibitor of bFGF activity. To better define the role of the intact endothelium in modulating the vascular and tissue deposition of bFGF, we compared the fate of intravenous injections of 125I-labeled bFGF with perivascular controlled growth factor release. Peak serum bFGF levels were detected within 1 min of injection, and the growth factor was cleared thereafter with a serum half-life of almost 3 min. Polymeric controlled release devices delivered bFGF to the extravascular space without transendothelial transport. Deposition within the blood vessel wall was rapidly distributed circumferentially and was substantially greater than that observed following intravenous injection. The amount of bFGF deposited in arteries adjacent to the release devices was 40 times that deposited in similar arteries in animals who received a single intravenous bolus of bFGF. Endothelial denudation had a minimal effect on deposition following perivascular release, and it increased deposition following intravenous delivery 2-fold. The presence of intimal hyperplasia increased deposition of perivascularly released bFGF 2.4-fold but decreased the deposition of intravenously injected bFGF by 67%. In contrast, bFGF was 5- to 30-fold more abundant in solid organs after intravenous injection than it was following perivascular release. Deposition was greatest in the kidney, liver, and spleen and was substantially lower in the heart and lung. Thus, bFGF is rapidly cleared following intravenous injection and is deposited within both solid organs and the walls of blood vessels. Unlike the mitogenic potential of bFGF within blood vessels, vascular deposition is virtually independent of the presence of endothelium. Perivascular delivery is far more efficient than intravenous delivery at depositing bFGF within the arterial wall, and an increased neointima may provide added substrate for potential bFGF deposition but has limited contact with intravascular growth factor as a result of dilutional and flow-mediated effects.
Collapse
Affiliation(s)
- E R Edelman
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA 02115
| | | | | |
Collapse
|
37
|
Murugesan G, Chisolm GM, Fox PL. Oxidized low density lipoprotein inhibits the migration of aortic endothelial cells in vitro. J Cell Biol 1993; 120:1011-9. [PMID: 8432723 PMCID: PMC2200083 DOI: 10.1083/jcb.120.4.1011] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Endothelial cell (EC) migration is a critical and initiating event in the formation of new blood vessels and in the repair of injured vessels. Compelling evidence suggests that oxidized low density lipoprotein (LDL) is present in atherosclerotic lesions, but its role in lesion formation has not been defined. We have examined the role of oxidized LDL in regulating the wound-healing response of vascular EC in vitro. Confluent cultures of bovine aortic EC were "wounded" with a razor, and migration was measured after 18 to 24 h as the number of cells moving into the wounded area and the mean distance of cells from the wound edge. Oxidized LDL markedly reduced migration in a concentration- and oxidation-dependent manner. Native LDL or oxidized LDL with a thiobarbituric acid (TBA) reactivity < 5 nmol malondialdehyde equivalents/mg cholesterol was not inhibitory; however, oxidized LDL with a TBA reactivity of 8-12 inhibited migration by 75-100%. Inhibition was half-maximal at 250-300 micrograms cholesterol/ml and nearly complete at 350-400 micrograms/ml. The antimigratory activity was not due to cell death since it was completely reversed 16 h after removal of the lipoprotein. The inhibitor molecule was shown to be a lipid; organic solvent extracts of oxidized LDL inhibited migration to nearly the same extent as the intact particle. When LDL was variably oxidized by dialysis against FeSO4 or CuSO4, or by UV irradiation, the inhibitory activity correlated with TBA reactivity and total lipid peroxides, but not with electrophoretic mobility or fluorescence (360 ex/430 em). This indicates that a lipid hydroperoxide may be the active species. These results suggest the possibility that oxidized LDL may limit the healing response of the endothelium after injury.
Collapse
Affiliation(s)
- G Murugesan
- Department of Cell Biology, Cleveland Clinic Research Institute, Ohio 44195
| | | | | |
Collapse
|
38
|
Edelman ER, Nugent MA, Smith LT, Karnovsky MJ. Basic fibroblast growth factor enhances the coupling of intimal hyperplasia and proliferation of vasa vasorum in injured rat arteries. J Clin Invest 1992; 89:465-73. [PMID: 1371124 PMCID: PMC442874 DOI: 10.1172/jci115607] [Citation(s) in RCA: 193] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Basic fibroblast growth factor (bFGF) is mitogenic for smooth muscle cells (SMC) and angiogenic. We examined the in vivo effects of bFGF in balloon denuded carotid arteries of laboratory rats. bFGF was administered continuously from polymer-based devices at 34 ng/d into the periadventitial space of rat carotid arteries for 2 wk. Intimal hyperplasia was not observed in the absence of injury or with lipopolysaccharide induced endothelial dysfunction. Different degrees of vascular injury produced proportionally more intimal hyperplasia. bFGF increased the intimal hyperplastic response 1.3-fold with severe vascular injury, and 2.4-fold with more mild injury. Increased cell proliferation, not extracellular matrix production, accounted for these effects. Cell density was unchanged for the control and bFGF-treated groups, and the number of proliferating intimal cells at 2 wk rose to an amount equivalent to the increase in mass; 1.9- and 4.0-fold for severe and lesser injury, respectively. The relative ability of heparin to reduce SMC proliferation was not altered by the presence of bFGF.bFGF also induced profound angiogenesis within and surrounding the polymeric releasing device, and in the vasa vasorum immediately around the injured arteries. bFGF's effect on vasa was linearly related to the amount of SMC proliferation within the blood vessel. Thus, the in vivo mitogenic and angiogenic potential of bFGF are coupled, and may be similarly modulated by the products of local injury and/or factors in the vessel wall.
Collapse
Affiliation(s)
- E R Edelman
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | | | | | | |
Collapse
|
39
|
Hayward P, Hokanson J, Heggers J, Fiddes J, Klingbeil C, Goeger M, Robson M. Fibroblast growth factor reserves the bacterial retardation of wound contraction. Am J Surg 1992; 163:288-93. [PMID: 1539760 DOI: 10.1016/0002-9610(92)90004-b] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chronic granulating wounds were established in rats by excising burns inoculated with Escherichia coli. Recombinant human basic fibroblast growth factor was applied at dosages of 1, 10, and 100 micrograms/cm2 to the wounds of three groups of 20 animals on days 5, 9, 12, 15, and 18 after injury. The rate of wound closure was compared with that of similarly wounded animals treated with saline vehicle alone. High levels of bacteria caused significant retardation of wound contraction. The addition of basic fibroblast growth factor at the 100 micrograms/cm2 dosage level markedly improved the rate of wound closure whereas inert vehicles applied alone were ineffective. Since bacterial counts did not decrease in the basic fibroblast growth factor treated wounds, basic fibroblast growth factor was not inherently bactericidal. Histologic examination of the wounds treated with basic fibroblast growth factor showed increased cellularity with increased numbers of fibroblasts and round cells. These results suggest basic fibroblast growth factor can overcome the defect in healing created by bacterial infection, and this peptide may have efficacy in the management of the contaminated wound.
Collapse
Affiliation(s)
- P Hayward
- Division of Plastic Surgery, University of Texas Medical Branch, Galveston 77550
| | | | | | | | | | | | | |
Collapse
|
40
|
Augustin-Voss HG, Pauli BU. Quantitative analysis of autocrine-regulated, matrix-induced, and tumor cell-stimulated endothelial cell migration using a silicon template compartmentalization technique. Exp Cell Res 1992; 198:221-7. [PMID: 1729131 DOI: 10.1016/0014-4827(92)90374-h] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Autocrine-regulated, matrix-induced, and tumor cell-stimulated endothelial cell migration was quantitatively analyzed using a two-dimensional, two-compartment coculture system. Silicon templates were used to subdivide 35-mm tissue culture dishes into two separate compartments. Endothelial cells were grown to confluence in the inner compartment and released from growth arrest by removal of the silicon template. The distance of endothelial cell outgrowth from the monolayer was measured in 24-h intervals. Endothelial cells from different vascular beds migrated with different migration rates (large vessel endothelial cells greater than hemangioendothelioma cells greater than microvessel endothelial cells). Prior coating of tissue culture wells with fibronectin, type I collagen, or type IV collagen and increasing serum concentrations strongly enhanced endothelial cell migration. Seeding tumor cells into the outer compartment prior to removal of the silicon template permitted the direct coculture analysis of tumor cell-induced endothelial cell migration. Microvascular endothelial cell migration was stimulated in a tumor cell number-dependent fashion, whereas large vessel endothelial cells could not consistently be stimulated by coculture with tumor cells. It is concluded that silicon templates offer a useful approach for the quantitative study of migration of anchorage-dependent cells, permitting follow-up measurements over several days, the study of matrix effects, and the direct coculture analysis of cell migration.
Collapse
Affiliation(s)
- H G Augustin-Voss
- Department of Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York 14853-6401
| | | |
Collapse
|
41
|
Affiliation(s)
- C Basilico
- Department of Microbiology, New York University School of Medicine, New York 10016
| | | |
Collapse
|
42
|
Malecaze F, Mathis A, Arné JL, Raulais D, Courtois Y, Hicks D. Localization of acidic fibroblast growth factor in proliferative vitreoretinopathy membranes. Curr Eye Res 1991; 10:719-29. [PMID: 1717200 DOI: 10.3109/02713689109013866] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The pathogenesis of proliferative vitreoretinopathy (PVR) membranes remains poorly understood. We have studied the presence of acidic fibroblast growth factor (aFGF), a potent mitogen for many cells, within these membranes. We have used affinity purified monospecific anti-aFGF polyclonal antibodies, in conjunction with highly sensitive immunofluorescence techniques. The labelling was exclusively localized to cell bodies and was absent from the extracellular matrix. Double labelling techniques revealed that all cytokeratin positive cells (probably pigmented epithelial cells) and macrophages contained aFGF-like immunoreactivity, whilst glial cells were unlabelled. Appropriate controls indicated the specificity of the antibodies. Hence, the presence of this mitogenic molecule within certain cell types constituting PVR membranes may contribute to the pathogenesis.
Collapse
Affiliation(s)
- F Malecaze
- Laboratoire d'Ophtalmologie, Hôpital de Rangueil, Toulouse, France
| | | | | | | | | | | |
Collapse
|
43
|
Yayon A, Klagsbrun M. Autocrine regulation of cell growth and transformation by basic fibroblast growth factor. Cancer Metastasis Rev 1990; 9:191-202. [PMID: 2292136 DOI: 10.1007/bf00046360] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Basic FGF (bFGF) and acidic FGF (aFGF) are multipotential factors that stimulate and support proliferation, migration and differentiation. Both bFGF and aFGF are non-secreted growth factors consistent with the lack of a signal peptide. However, bFGF and aFGF are deposited in extracellular matrix (ECM) suggesting that an alternative mechanism for FGF release exists. Four oncogenes, int-2, hst/K-fgf, FGF-5 and FGF-6 have been isolated that are highly homologous to aFGF and bFGF. Unlike bFGF and aFGF, they possess signal peptides and are secreted. These oncogenes transform cells and induce tumors, ostensibly via an autocrine mechanism. The involvement of bFGF and aFGF in autocrine transformation has been clarified by studies using FGF cDNA transfection. NIH-3T3 cells transfected with native bFGF cDNA and expressing 20 to 100 times as much bFGF as parental 3T3 cells acquire an enhanced proliferation rate and higher saturation density. NIH cells transfected with a construct in which bFGF cDNA is altered by addition of a signal peptide, undergo autocrine transformation and exhibit morphological and biochemical alterations characteristic of highly transformed cells. Injection of cells expressing native bFGF even at levels 100 times greater than parental 3T3 cells fails to induce tumors or lung metastasis in syngeneic mice. Signal peptide bFGF-transected cells on the other hand, acquire a high tumorigenic and metastatic potential with tumor incidence and numbers comparable to those induced by ras transformed cells. Acquisition of a signal peptide converts bFGF into a transforming protein analogous to FGF-related oncogenes which naturally have signal peptide sequences.
Collapse
Affiliation(s)
- A Yayon
- Department of Surgery, Children's Hospital, Boston, MA 02115
| | | |
Collapse
|
44
|
Anagnostou A, Lee ES, Kessimian N, Levinson R, Steiner M. Erythropoietin has a mitogenic and positive chemotactic effect on endothelial cells. Proc Natl Acad Sci U S A 1990; 87:5978-82. [PMID: 2165612 PMCID: PMC54453 DOI: 10.1073/pnas.87.15.5978] [Citation(s) in RCA: 434] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Erythropoietin is known to be a hematopoietic growth factor with a singularly specific action on the proliferation and differentiation of erythroid progenitor cells. We have observed a dose-dependent proliferative action of human recombinant erythropoietin on human umbilical vein endothelial cells and bovine adrenal capillary endothelial cells. Binding studies with radioiodinated recombinant human erythropoietin revealed a large number (approximately 27,000) of an apparent single class of receptors with an affinity in the 10(-9) M range. Linkage of the radiolabeled ligand to its receptor via a bifunctional crosslinking agent allowed us to identify an endothelial cell protein of 45 kDa as the principal receptor associated with this mitogenic effect of erythropoietin. Recombinant human erythropoietin also enhanced the migration of endothelial cells.
Collapse
Affiliation(s)
- A Anagnostou
- Department of Medicine Memorial Hospital of Rhode Island, Pawtucket 02860
| | | | | | | | | |
Collapse
|
45
|
Shoji S, Ertl RF, Linder J, Koizumi S, Duckworth WC, Rennard SI. Bronchial epithelial cells respond to insulin and insulin-like growth factor-I as a chemoattractant. Am J Respir Cell Mol Biol 1990; 2:553-7. [PMID: 2189458 DOI: 10.1165/ajrcmb/2.6.553] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Migration of epithelial cells to cover areas of injury is thought to be important in the repair process following airway insult. Insulin is reported to be a growth factor for bronchial epithelial cells, and growth factors have been known to be chemotactic for many types of cells. Thus, we hypothesized that insulin may be a chemoattractant for bronchial epithelial cells. To evaluate this, we prepared bronchial epithelial cells and measured their chemotactic activity toward insulin. Bronchial epithelial cells were isolated by overnight digestion with bacterial protease, filtered through 100-microns nitex mesh, and then cultured at 1 x 10(6) cells/ml in tissue culture dishes in medium 199 supplemented with transferrin, insulin, epidermal growth factor, hydrocortisone, antibiotics, and 10% FCS for 3 d. The cultured cells were rinsed twice to remove supplements, trypsinized and resuspended at 1 x 10(6) cells/ml in medium 199 without supplements, and used as the cell source for chemotaxis. Chemotactic activity of bronchial epithelial cells was measured by the blindwell chamber technique using 8-microns Nuclepore filter membranes coated with 0.1% gelatin. The cells were added to the top wells in a 48-multiwell chamber with insulin in the bottom wells and incubated for 6 h at 37 degrees C, 5% CO2. Bronchial epithelial cells migrated in response to insulin in a dose-dependent manner up to an optimal dose of insulin, 100 micrograms/ml, and decreased at higher concentrations. The number of migrated cells per 10 high power fields was 33.7 +/- 1.9 at the optimum and 3.7 +/- 0.7 without insulin (P less than 0.01).(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- S Shoji
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha 68198-2465
| | | | | | | | | | | |
Collapse
|
46
|
Rosen EM, Goldberg ID. Protein factors which regulate cell motility. IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY : JOURNAL OF THE TISSUE CULTURE ASSOCIATION 1989; 25:1079-87. [PMID: 2558106 DOI: 10.1007/bf02621258] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell motility (i.e., movement) is an essential component of normal development, inflammation, tissue repair, angiogenesis, and tumor invasion. Various molecules can affect the motility and positioning of mammalian cells, including peptide growth factors, (e.g., EGF, PDGF, TGF-beta), substrate-adhesion molecules (e.g., fibronectin, laminin), cell adhesion molecules (CAMs), and metalloproteinases. Recent studies have demonstrated a group of motility-stimulating proteins which do not appear to fit into any of the above categories. Examples include: 1) scatter factor (SF), a mesenchymal cell-derived protein which causes contiguous sheets of epithelium to separate into individual cells and stimulates the migration of epithelial as well as vascular endothelial cells; 2) autocrine motility factor (AMF), a tumor cell-derived protein which stimulates migration of the producer cells; and 3) migration-stimulating factor (MSF), a protein produced by fetal and cancer patient fibroblasts which stimulates penetration of three-dimensional collagen gels by non-producing adult fibroblasts. SF, AMF, and MSF are soluble and heat labile proteins with Mr of 77, 55, and 70 kd by SDS-PAGE, respectively, and may be members of a new class of cell-specific regulators of motility. Their physiologic functions have not been established, but available data suggest that they may be involved in fetal development and/or tissue repair.
Collapse
Affiliation(s)
- E M Rosen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06510
| | | |
Collapse
|
47
|
Long MM, King VJ, Prasad KU, Freeman BA, Urry DW. Elastin repeat peptides as chemoattractants for bovine aortic endothelial cells. J Cell Physiol 1989; 140:512-8. [PMID: 2476449 DOI: 10.1002/jcp.1041400316] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cultured bovine aortic endothelial cells migrate toward a concentration gradient of repeating elastin peptides, specifically the repeating nonamers Gly-Phe-Gly-Val-Gly-Ala-Gly-Val-Pro and Gly-Leu-Gly-Val-Gly-Ala-Gly-Val-Pro and the repeating hexamer Val-Gly-Val-Ala-Pro-Gly. Dose-response experiments demonstrate that the peak of activity occurs at 8 x 10(-8) M for the nonapeptides and 1 x 10(-8) M for the hexapeptide. Checkerboard assays establish that the movement is chemotaxis and not chemokinesis. Because of the concentration difference in the responsiveness between the nonapeptide and the hexapeptide, the cells can differentiate between the two types of repeats. The positive control for the chemotaxis studies was fibronectin.
Collapse
Affiliation(s)
- M M Long
- Department of Anesthesiology, School of Medicine, University of Alabama, Birmingham 35294
| | | | | | | | | |
Collapse
|
48
|
Kurokawa M, Doctrow SR, Klagsbrun M. Neutralizing antibodies inhibit the binding of basic fibroblast growth factor to its receptor but not to heparin. J Biol Chem 1989. [DOI: 10.1016/s0021-9258(18)83289-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
49
|
Klagsbrun M, Edelman ER. Biological and biochemical properties of fibroblast growth factors. Implications for the pathogenesis of atherosclerosis. ARTERIOSCLEROSIS (DALLAS, TEX.) 1989; 9:269-78. [PMID: 2655570 DOI: 10.1161/01.atv.9.3.269] [Citation(s) in RCA: 186] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- M Klagsbrun
- Department of Biological Chemistry, Children's Hospital, Boston, MA 02115
| | | |
Collapse
|
50
|
Shipley GD, Keeble WW, Hendrickson JE, Coffey RJ, Pittelkow MR. Growth of normal human keratinocytes and fibroblasts in serum-free medium is stimulated by acidic and basic fibroblast growth factor. J Cell Physiol 1989; 138:511-8. [PMID: 2466852 DOI: 10.1002/jcp.1041380310] [Citation(s) in RCA: 127] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Keratinocytes and fibroblasts isolated from human neonatal foreskin can be plated and grown through multiple rounds of division in vitro under defined serum-free conditions. We utilized these growth conditions to examine the mitogenic potential of acidic and basic fibroblast growth factor (aFGF and bFGF) on these cells. Our results demonstrate that both aFGF and bFGF can stimulate the proliferation of keratinocytes and fibroblasts. aFGF is a more potent mitogen than bFGF for keratinocytes. In contrast, bFGF appears to be more potent than aFGF in stimulating the growth of fibroblast cultures. Heparin sulfate (10 micrograms/ml) dramatically inhibited the ability of bFGF to stimulate the proliferation of keratinocytes. In comparison, heparin slightly inhibited the stimulatory effect of aFGF and had no effect on epidermal growth factor (EGF) stimulation in keratinocyte cultures. In fibroblast cultures the addition of heparin enhanced the mitogenic effect of aFGF, had a minimal stimulatory effect on the mitogenic activity of bFGF, and had no effect on EGF-stimulated growth. Our results demonstrate that the proliferation in vitro of two normal cell types found in the skin can be influenced by aFGF and bFGF and demonstrate cell-type specific differences in the responsiveness of fibroblasts and keratinocytes to these growth factors and heparin.
Collapse
Affiliation(s)
- G D Shipley
- Department of Cell Biology and Anatomy, School of Medicine, Oregon Health Sciences University, Portland 97201
| | | | | | | | | |
Collapse
|