1
|
Prakash R, Izraely S, Thareja NS, Lee RH, Rappaport M, Kawaguchi R, Sagi-Assif O, Ben-Menachem S, Meshel T, Machnicki M, Ohe S, Hoon DS, Coppola G, Witz IP, Carmichael ST. Regeneration Enhances Metastasis: A Novel Role for Neurovascular Signaling in Promoting Melanoma Brain Metastasis. Front Neurosci 2019; 13:297. [PMID: 31024232 PMCID: PMC6465799 DOI: 10.3389/fnins.2019.00297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
Neural repair after stroke involves initiation of a cellular proliferative program in the form of angiogenesis, neurogenesis, and molecular growth signals in the surrounding tissue elements. This cellular environment constitutes a niche in which regeneration of new blood vessels and new neurons leads to partial tissue repair after stroke. Cancer metastasis has similar proliferative cellular events in the brain and other organs. Do cancer and CNS tissue repair share similar cellular processes? In this study, we identify a novel role of the regenerative neurovascular niche induced by stroke in promoting brain melanoma metastasis through enhancing cellular interactions with surrounding niche components. Repair-mediated neurovascular signaling induces metastatic cells to express genes crucial to metastasis. Mimicking stroke-like conditions in vitro displays an enhancement of metastatic migration potential and allows for the determination of cell-specific signals produced by the regenerative neurovascular niche. Comparative analysis of both in vitro and in vivo expression profiles reveals a major contribution of endothelial cells in mediating melanoma metastasis. These results point to a previously undiscovered role of the regenerative neurovascular niche in shaping the tumor microenvironment and brain metastatic landscape.
Collapse
Affiliation(s)
- Roshini Prakash
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sivan Izraely
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nikita S Thareja
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rex H Lee
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maya Rappaport
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Riki Kawaguchi
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Orit Sagi-Assif
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shlomit Ben-Menachem
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Machnicki
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shuichi Ohe
- Department of Translational Molecular Medicine, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Dave S Hoon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isaac P Witz
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
2
|
Das A, Ghatak S, Sinha M, Chaffee S, Ahmed NS, Parinandi NL, Wohleb ES, Sheridan JF, Sen CK, Roy S. Correction of MFG-E8 Resolves Inflammation and Promotes Cutaneous Wound Healing in Diabetes. THE JOURNAL OF IMMUNOLOGY 2016; 196:5089-100. [PMID: 27194784 DOI: 10.4049/jimmunol.1502270] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
Milk fat globule epidermal growth factor-factor 8 (MFG-E8) is a peripheral glycoprotein that acts as a bridging molecule between the macrophage and apoptotic cells, thus executing a pivotal role in the scavenging of apoptotic cells from affected tissue. We have previously reported that apoptotic cell clearance activity or efferocytosis is compromised in diabetic wound macrophages. In this work, we test the hypothesis that MFG-E8 helps resolve inflammation, supports angiogenesis, and accelerates wound closure. MFG-E8(-/-) mice displayed impaired efferocytosis associated with exaggerated inflammatory response, poor angiogenesis, and wound closure. Wound macrophage-derived MFG-E8 was recognized as a critical driver of wound angiogenesis. Transplantation of MFG-E8(-/-) bone marrow to MFG-E8(+/+) mice resulted in impaired wound closure and compromised wound vascularization. In contrast, MFG-E8(-/-) mice that received wild-type bone marrow showed improved wound closure and improved wound vascularization. Hyperglycemia and exposure to advanced glycated end products inactivated MFG-E8, recognizing a key mechanism that complicates diabetic wound healing. Diabetic db/db mice suffered from impaired efferocytosis accompanied with persistent inflammation and slow wound closure. Topical recombinant MFG-E8 induced resolution of wound inflammation, improvements in angiogenesis, and acceleration of closure, upholding the potential of MFG-E8-directed therapeutics in diabetic wound care.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Subhadip Ghatak
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Mithun Sinha
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Scott Chaffee
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Noha S Ahmed
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Narasimham L Parinandi
- Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Eric S Wohleb
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - Chandan K Sen
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Sashwati Roy
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210;
| |
Collapse
|
3
|
Furusawa K, Mizutani T, Sasaki N. Development of the evaluation system for barrier functions of engineered epithelial lumens. Regen Ther 2016; 3:82-89. [PMID: 31245477 PMCID: PMC6581833 DOI: 10.1016/j.reth.2016.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 11/18/2022] Open
Abstract
We have investigated the effects of a diameter of engineered epithelial lumen on cellar architectures and a barrier function. For this investigation, we have developed a system to evaluate the barrier function of engineered epithelial lumens. To test the utility of our system, we constructed the engineered epithelial lumens by culturing Madin-Darby Canine Kidney cells (MDCK) on the gold wires with different diameters ranging from 50 μm-200 μm. Confocal laser scanning microscopy revealed that long actin stress fibers and a low focal adhesion density were observed at the gold wire diameter of 200 μm, whereas the mesh-like morphology consisted of short actin stress fibers and high focal adhesion densities were found at the gold wire diameters of 50 μm and 100 μm. The expression pattern of ZO-1 that localizes at the tight junction was independent on the gold wire diameter. The electrical impedance measurement indicates that the barrier function for the samples constructed at the gold wire diameter of 200 μm was significantly higher than those at the gold wire diameters of 50 μm and 100 μm. The difference in the barrier functions of epithelial lumens might be attributed to the changes in cellular architectures with increasing the curvature of gold wire.
Collapse
Affiliation(s)
- Kazuya Furusawa
- Faculty of Advanced Life Science, Hokkaido University, Kita-ku, Kita 10 Nishi 8, Sapporo, Hokkaido, Japan
| | | | | |
Collapse
|
4
|
Liao WY, Li HJ, Chang MY, Tang ACL, Hoffman AS, Hsieh PCH. Comprehensive characterizations of nanoparticle biodistribution following systemic injection in mice. NANOSCALE 2013; 5:11079-11086. [PMID: 24072256 DOI: 10.1039/c3nr03954d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Various nanoparticle (NP) properties such as shape and surface charge have been studied in an attempt to enhance the efficacy of NPs in biomedical applications. When trying to undermine the precise biodistribution of NPs within the target organs, the analytical method becomes the determining factor in measuring the precise quantity of distributed NPs. High performance liquid chromatography (HPLC) represents a more powerful tool in quantifying NP biodistribution compared to conventional analytical methods such as an in vivo imaging system (IVIS). This, in part, is due to better curve linearity offered by HPLC than IVIS. Furthermore, HPLC enables us to fully analyze each gram of NPs present in the organs without compromising the signals and the depth-related sensitivity as is the case in IVIS measurements. In addition, we found that changing physiological conditions improved large NP (200-500 nm) distribution in brain tissue. These results reveal the importance of selecting analytic tools and physiological environment when characterizing NP biodistribution for future nanoscale toxicology, therapeutics and diagnostics.
Collapse
Affiliation(s)
- Wei-Yin Liao
- Institute of Clinical Medicine, National Cheng Kung University & Hospital, Tainan 704, Taiwan, R.O.C..
| | | | | | | | | | | |
Collapse
|
5
|
A PPARγ-FGF1 axis is required for adaptive adipose remodelling and metabolic homeostasis. Nature 2012; 485:391-4. [PMID: 22522926 PMCID: PMC3358516 DOI: 10.1038/nature10998] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 02/27/2012] [Indexed: 12/28/2022]
Abstract
Although feast and famine cycles illustrate that remodelling of adipose tissue in response to fluctuations in nutrient availability is essential for maintaining metabolic homeostasis, the underlying mechanisms remain poorly understood. Here we identify fibroblast growth factor 1 (FGF1) as a critical transducer in this process in mice, and link its regulation to the nuclear receptor PPARγ (peroxisome proliferator activated receptor γ), which is the adipocyte master regulator and the target of the thiazolidinedione class of insulin sensitizing drugs. FGF1 is the prototype of the 22-member FGF family of proteins and has been implicated in a range of physiological processes, including development, wound healing and cardiovascular changes. Surprisingly, FGF1 knockout mice display no significant phenotype under standard laboratory conditions. We show that FGF1 is highly induced in adipose tissue in response to a high-fat diet and that mice lacking FGF1 develop an aggressive diabetic phenotype coupled to aberrant adipose expansion when challenged with a high-fat diet. Further analysis of adipose depots in FGF1-deficient mice revealed multiple histopathologies in the vasculature network, an accentuated inflammatory response, aberrant adipocyte size distribution and ectopic expression of pancreatic lipases. On withdrawal of the high-fat diet, this inflamed adipose tissue fails to properly resolve, resulting in extensive fat necrosis. In terms of mechanisms, we show that adipose induction of FGF1 in the fed state is regulated by PPARγ acting through an evolutionarily conserved promoter proximal PPAR response element within the FGF1 gene. The discovery of a phenotype for the FGF1 knockout mouse establishes the PPARγ–FGF1 axis as critical for maintaining metabolic homeostasis and insulin sensitization.
Collapse
|
6
|
Roche B, David V, Vanden-Bossche A, Peyrin F, Malaval L, Vico L, Lafage-Proust MH. Structure and quantification of microvascularisation within mouse long bones: what and how should we measure? Bone 2012; 50:390-9. [PMID: 22019874 DOI: 10.1016/j.bone.2011.09.051] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 09/07/2011] [Accepted: 09/09/2011] [Indexed: 10/16/2022]
Abstract
Bone marrow vascularisation is involved in both remodeling and hematopoïesis. Challenged mouse models often require imaging and quantitative assessment of blood vessels and bone cell activities for a better understanding of the role of the vascular system. In this study we compared images of mouse hind limb long bone vascularisation after infusion of either barium sulfate or lead chromate-loaded silicon. The images were then analyzed through histology as well as low-resolution and synchrotron-radiation microtomography. We show that barium sulfate infusion provides the best vessel images and furthermore, that it is compatible with staining procedures used in bone histomorphometry and CD31 immunohistochemistry. Bone marrow vascularisation displays large structural and spatial distribution heterogeneity, including large lobular clusters of sinusoids and an unexpectedly substantial amount of capillaries in the adipocytes-rich distal third of the tibia. For an unbiased assessment of bone vascular development/changes, these features must be taken into account. We describe the conditions under which the quantification of microvascularisation on histological sections of barium-infused long bones is reproducible, as applied to seven-month-old male C57/Bl6J and mixed CD1/129Sv/J mice, and we propose a nomenclature for the histological parameters measured. Finally, we validate our technique by studying the effect of ovariectomy on mouse tibial vascular density.
Collapse
|
7
|
Qiao L, Lu SL, Dong JY, Song F. Abnormal regulation of neo-vascularisation in deep partial thickness scalds in rats with diabetes mellitus. Burns 2011; 37:1015-22. [DOI: 10.1016/j.burns.2011.03.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 03/27/2011] [Accepted: 03/29/2011] [Indexed: 11/15/2022]
|
8
|
Kim HA, Rhim T, Lee M. Regulatory systems for hypoxia-inducible gene expression in ischemic heart disease gene therapy. Adv Drug Deliv Rev 2011; 63:678-87. [PMID: 21241757 DOI: 10.1016/j.addr.2011.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 12/29/2010] [Accepted: 01/05/2011] [Indexed: 12/30/2022]
Abstract
Ischemic heart diseases are caused by narrowed coronary arteries that decrease the blood supply to the myocardium. In the ischemic myocardium, hypoxia-responsive genes are up-regulated by hypoxia-inducible factor-1 (HIF-1). Gene therapy for ischemic heart diseases uses genes encoding angiogenic growth factors and anti-apoptotic proteins as therapeutic genes. These genes increase blood supply into the myocardium by angiogenesis and protect cardiomyocytes from cell death. However, non-specific expression of these genes in normal tissues may be harmful, since growth factors and anti-apoptotic proteins may induce tumor growth. Therefore, tight gene regulation is required to limit gene expression to ischemic tissues, to avoid unwanted side effects. For this purpose, various gene expression strategies have been developed for ischemic-specific gene expression. Transcriptional, post-transcriptional, and post-translational regulatory strategies have been developed and evaluated in ischemic heart disease animal models. The regulatory systems can limit therapeutic gene expression to ischemic tissues and increase the efficiency of gene therapy. In this review, recent progresses in ischemic-specific gene expression systems are presented, and their applications to ischemic heart diseases are discussed.
Collapse
|
9
|
Narkar VA, Fan W, Downes M, Yu RT, Jonker JW, Alaynick WA, Banayo E, Karunasiri MS, Lorca S, Evans RM. Exercise and PGC-1α-independent synchronization of type I muscle metabolism and vasculature by ERRγ. Cell Metab 2011; 13:283-93. [PMID: 21356518 PMCID: PMC3084588 DOI: 10.1016/j.cmet.2011.01.019] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 06/03/2010] [Accepted: 01/26/2011] [Indexed: 01/23/2023]
Abstract
How type I skeletal muscle inherently maintains high oxidative and vascular capacity in the absence of exercise is unclear. We show that nuclear receptor ERRγ is highly expressed in type I muscle and, when transgenically expressed in anaerobic type II muscles (ERRGO mice), dually induces metabolic and vascular transformation in the absence of exercise. ERRGO mice show increased expression of genes promoting fat metabolism, mitochondrial respiration, and type I fiber specification. Muscles in ERRGO mice also display an activated angiogenic program marked by myofibrillar induction and secretion of proangiogenic factors, neovascularization, and a 100% increase in running endurance. Surprisingly, the induction of type I muscle properties by ERRγ does not involve PGC-1α. Instead, ERRγ genetically activates the energy sensor AMPK in mediating the metabovascular changes in ERRGO mice. Therefore, ERRγ represents a previously unrecognized determinant that specifies intrinsic vascular and oxidative metabolic features that distinguish type I from type II muscle.
Collapse
Affiliation(s)
- Vihang A. Narkar
- Gene Expression Laboratory, Salk Institute, La Jolla, California
| | | | - Michael Downes
- Gene Expression Laboratory, Salk Institute, La Jolla, California
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute, La Jolla, California
| | - Johan W. Jonker
- Gene Expression Laboratory, Salk Institute, La Jolla, California
| | | | - Ester Banayo
- Gene Expression Laboratory, Salk Institute, La Jolla, California
| | | | | | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute, La Jolla, California
- Howard Hughes Medical Institute, La Jolla, California
| |
Collapse
|
10
|
Targeted in vivo extracellular matrix formation promotes neovascularization in a rodent model of myocardial infarction. PLoS One 2010; 5:e10384. [PMID: 20442783 PMCID: PMC2860995 DOI: 10.1371/journal.pone.0010384] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 04/07/2010] [Indexed: 12/22/2022] Open
Abstract
Background The extracellular matrix plays an important role in tissue regeneration. We investigated whether extracellular matrix protein fragments could be targeted with antibodies to ischemically injured myocardium to promote angiogenesis and myocardial repair. Methodology/Principal Findings Four peptides, 2 derived from fibronectin and 2 derived from Type IV Collagen, were assessed for in vitro and in vivo tendencies for angiogenesis. Three of the four peptides—Hep I, Hep III, RGD—were identified and shown to increase endothelial cell attachment, proliferation, migration and cell activation in vitro. By chemically conjugating these peptides to an anti-myosin heavy chain antibody, the peptides could be administered intravenously and specifically targeted to the site of the myocardial infarction. When administered into Sprague-Dawley rats that underwent ischemia-reperfusion myocardial infarction, these peptides produced statistically significantly higher levels of angiogenesis and arteriogenesis 6 weeks post treatment. Conclusions/Significance We demonstrated that antibody-targeted ECM-derived peptides alone can be used to sufficiently alter the extracellular matrix microenvironment to induce a dramatic angiogenic response in the myocardial infarct area. Our results indicate a potentially new non-invasive strategy for repairing damaged tissue, as well as a novel tool for investigating in vivo cell biology.
Collapse
|
11
|
Abstract
Therapies that aim to prevent myocardial tissue from dying or to regenerate new myocardium all rely on the preservation or growth of a functional vasculature. The amount of blood that supplies the myocardium is dependent on the number and nature of the microvessels, as well as the ability of the arteries to supply blood and the veins to remove it. All of these factors can be assessed when success of an experimental therapy is being evaluated. Different kinds of information can be obtained from these different parameters, and it is important to understand what each one involves and how it can be misinterpreted. This chapter describes the various approaches to the assessment of vascularity in the heart with a focus on small animal models, dealing both with those approaches that are purely histological endpoint studies and those that are functional measurements in living animals.
Collapse
|
12
|
Abramyuk A, Tokalov SV. Distribution of fluorescent microspheres in vascular space and parenchymal organs of intact nude rats. Int J Radiat Biol 2009; 85:781-6. [PMID: 19657864 DOI: 10.1080/09553000903090035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE To assess kinetics of elimination of different sized microspheres (MS) from the blood pool and tendency of their distribution in parenchymal organs of intact nude rats. MATERIALS AND METHODS A mixture of 1 microm and 3 microm MS in phosphate-buffered saline was injected intravenously into eight rats under intraperitoneal anaesthesia. Blood samples were collected before, just after and in 2, 5 and 10 min after MS injection. Dynamics of MS elimination from blood pool was evaluated with flow cytometry. After euthanasia, histological sections were prepared and distributions of MS through the liver, spleen, kidney and lung were analysed with fluorescence microscopy and flow cytometry. RESULTS The number of microspheres registered in the intravascular space showed a marked exponential decrease over time independent of MS size. Different amounts and proportions of 1 microm and 3 microm MS were revealed in lung, liver, spleen and kidneys of the rats. Most of 1 microm MS were localised in liver and spleen. In contrast, 3 microm MS were detected predominantly in lung. CONCLUSION 1 microm and 3 microm MS may be assumed as free circulating particles only for a short period of time after injection. Their elimination kinetics seems to be tightly linked to specific tissue properties such a pulmonary vasoconstriction and phagocytosis.
Collapse
Affiliation(s)
- Andrij Abramyuk
- OncoRay-Center for Radiation Research in Oncology, Dresden University of Technology, Dresden, Germany.
| | | |
Collapse
|
13
|
Integrin beta1-focal adhesion kinase signaling directs the proliferation of metastatic cancer cells disseminated in the lungs. Proc Natl Acad Sci U S A 2009; 106:10290-5. [PMID: 19502425 DOI: 10.1073/pnas.0904227106] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The development of metastases is an extended and inefficient process involving multiple steps. The last of these involves the growth of micrometastases into macroscopic tumors. We show here that intravenously injected, nonmetastatic cancer cells cease proliferating after extravasating into the parenchyma of the lungs; this response is attributable to the cells inability to trigger adhesion-related signaling events when they are scattered sparsely within the extracellular matrix (ECM) of the parenchyma. We recapitulate this situation by culturing these nonmetastatic cells at low seeding density in ECM-derived gels in vitro, in which they undergo cell-cycle arrest resulting, in part, from insufficient activation of focal adhesion kinase (FAK). Metastatic cancer cells, in contrast, show sufficient FAK activation to enable their proliferation within ECM gels in vitro and continue cell-cycle progression within the lung parenchyma in vivo. Activation of FAK in these metastatic cells depends on expression of the beta(1) subunit of integrins, and proliferation of these cells after extravasation in the lungs is diminished by knocking down the expression of either FAK or integrin beta(1). These results demonstrate the critical role of integrin beta(1)-FAK signaling axis in controlling the initial proliferation of micrometastatic cancer cells disseminated in the lungs.
Collapse
|
14
|
Hardy KM, Dillaman RM, Locke BR, Kinsey ST. A skeletal muscle model of extreme hypertrophic growth reveals the influence of diffusion on cellular design. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1855-67. [PMID: 19321701 DOI: 10.1152/ajpregu.00076.2009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Muscle fibers that power swimming in the blue crab Callinectes sapidus are <80 microm in diameter in juveniles but grow hypertrophically, exceeding 600 microm in adults. Therefore, intracellular diffusion distances become progressively greater as the animals grow and, in adults, vastly exceed those in most cells. This developmental trajectory makes C. sapidus an excellent model for characterization of the influence of diffusion on fiber structure. The anaerobic light fibers, which power burst swimming, undergo a prominent shift in organelle distribution with growth. Mitochondria, which require O2 and rely on the transport of small, rapidly diffusing metabolites, are evenly distributed throughout the small fibers of juveniles, but in the large fibers of adults they are located almost exclusively at the fiber periphery where O2 concentrations are high. Nuclei, which do not require O2, but rely on the transport of large, slow-moving macromolecules, have the inverse pattern: they are distributed peripherally in small fibers but are evenly distributed across the large fibers, thereby reducing diffusion path lengths for large macromolecules. The aerobic dark fibers, which power endurance swimming, have evolved an intricate network of cytoplasmically isolated, highly perfused subdivisions that create the short diffusion distances needed to meet the high aerobic ATP turnover demands of sustained contraction. However, fiber innervation patterns are the same in the dark and light fibers. Thus the dark fibers appear to have disparate functional units for metabolism (fiber subdivision) and contraction (entire fiber). Reaction-diffusion mathematical models demonstrate that diffusion would greatly constrain the rate of metabolic processes without these developmental changes in fiber structure.
Collapse
Affiliation(s)
- Kristin M Hardy
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC 28403-5915, USA
| | | | | | | |
Collapse
|
15
|
Aicher A, Heeschen C, Feil S, Hofmann F, Mendelsohn ME, Feil R, Dimmeler S. cGMP-dependent protein kinase I is crucial for angiogenesis and postnatal vasculogenesis. PLoS One 2009; 4:e4879. [PMID: 19287493 PMCID: PMC2654072 DOI: 10.1371/journal.pone.0004879] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2008] [Accepted: 02/10/2009] [Indexed: 01/13/2023] Open
Abstract
Background Endothelium-derived nitric oxide plays an important role for the bone marrow microenvironment. Since several important effects of nitric oxide are mediated by cGMP-dependent pathways, we investigated the role of the cGMP downstream effector cGMP-dependent protein kinase I (cGKI) on postnatal neovascularization. Methodology/Principal Findings In a disc neovascularization model, cGKI−/− mice showed an impaired neovascularization as compared to their wild-type (WT) littermates. Infusion of WT, but not cGKI−/− bone marrow progenitors rescued the impaired ingrowth of new vessels in cGKI-deficient mice. Bone marrow progenitors from cGKI−/− mice showed reduced proliferation and survival rates. In addition, we used cGKIα leucine zipper mutant (LZM) mice as model for cGKI deficiency. LZM mice harbor a mutation in the cGKIα leucine zipper that prevents interaction with downstream signaling molecules. Consistently, LZM mice exhibited reduced numbers of vasculogenic progenitors and impaired neovascularization following hindlimb ischemia compared to WT mice. Conclusions/Significance Our findings demonstrate that the cGMP-cGKI pathway is critical for postnatal neovascularization and establish a new role for cGKI in vasculogenesis, which is mediated by bone marrow-derived progenitors.
Collapse
Affiliation(s)
- Alexandra Aicher
- Department of Internal Medicine III, J. W. Goethe University, Frankfurt, Germany
- * E-mail: (AA); (SD)
| | - Christopher Heeschen
- Department of Internal Medicine III, J. W. Goethe University, Frankfurt, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, Universität Tübingen, Tübingen, Germany
| | - Franz Hofmann
- Institut für Pharmakologie und Toxikologie, Technische Universität München, München, Germany
| | - Michael E. Mendelsohn
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Robert Feil
- Interfakultäres Institut für Biochemie, Universität Tübingen, Tübingen, Germany
| | - Stefanie Dimmeler
- Department of Internal Medicine III, J. W. Goethe University, Frankfurt, Germany
- * E-mail: (AA); (SD)
| |
Collapse
|
16
|
Lactate stimulates vasculogenic stem cells via the thioredoxin system and engages an autocrine activation loop involving hypoxia-inducible factor 1. Mol Cell Biol 2008; 28:6248-61. [PMID: 18710947 DOI: 10.1128/mcb.00795-08] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The recruitment and differentiation of circulating stem/progenitor cells (SPCs) in subcutaneous Matrigel in mice was assessed. There were over one million CD34(+) SPCs per Matrigel plug 18 h after Matrigel implantation, and including a polymer to elevate the lactate concentration increased the number of SPCs by 3.6-fold. Intricate CD34(+) cell-lined channels were linked to the systemic circulation, and lactate accelerated cell differentiation as evaluated based on surface marker expression and cell cycle entry. CD34(+) SPCs from lactate-supplemented Matrigel exhibited significantly higher concentrations of thioredoxin 1 (Trx1) and hypoxia-inducible factor 1 (HIF-1) than cells from unsupplemented Matrigel, whereas Trx1 and HIF-1 in CD45(+) leukocytes were not elevated by lactate. Results obtained using small inhibitory RNA (siRNA) specific to HIF-1 and mice with conditionally HIF-1 null myeloid cells indicated that SPC recruitment and lactate-mediated effects were dependent on HIF-1. Cells from lactate-supplemented Matrigel had higher concentrations of phosphorylated extracellular signal-regulated kinases 1 and 2, Trx1, Trx reductase (TrxR), vascular endothelial growth factor (VEGF), and stromal cell-derived factor 1 (SDF-1) than cells from unsupplemented Matrigel. SPC recruitment and protein changes were inhibited by siRNA specific to lactate dehydrogenase, TrxR, or HIF-1 and by oxamate, apocynin, U0126, N-acetylcysteine, dithioerythritol, and antibodies to VEGF or SDF-1. Oxidative stress from lactate metabolism by SPCs accelerated further SPC recruitment and differentiation through Trx1-mediated elevations in HIF-1 levels and the subsequent synthesis of HIF-1-dependent growth factors.
Collapse
|
17
|
Cao L, Mooney DJ. Spatiotemporal control over growth factor signaling for therapeutic neovascularization. Adv Drug Deliv Rev 2007; 59:1340-50. [PMID: 17868951 PMCID: PMC2581871 DOI: 10.1016/j.addr.2007.08.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Accepted: 08/01/2007] [Indexed: 12/29/2022]
Abstract
Many of the qualitative roles of growth factors involved in neovascularization have been delineated, but it is unclear yet from an engineering perspective how to use these factors as therapies. We propose that an approach that integrates quantitative spatiotemporal measurements of growth factor signaling using 3-D in vitro and in vivo models, mathematic modeling of factor tissue distribution, and new delivery technologies may provide an opportunity to engineer neovascularization on demand.
Collapse
Affiliation(s)
| | - David J. Mooney
- Corresponding author. Harvard University, 319 Pierce Hall, 29 Oxford, Street, Cambridge, MA 02138, USA. Tel.: +1 617 384 9624; fax: +1 617 495 9837. E-mail address: (D.J. Mooney)
| |
Collapse
|
18
|
Breitbach CJ, Paterson JM, Lemay CG, Falls TJ, McGuire A, Parato KA, Stojdl DF, Daneshmand M, Speth K, Kirn D, McCart JA, Atkins H, Bell JC. Targeted inflammation during oncolytic virus therapy severely compromises tumor blood flow. Mol Ther 2007; 15:1686-93. [PMID: 17579581 DOI: 10.1038/sj.mt.6300215] [Citation(s) in RCA: 211] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Oncolytic viruses (OVs) are selected or designed to eliminate malignancies by direct infection and lysis of cancer cells. In contrast to this concept of direct tumor lysis by viral infection, we observed that a significant portion of the in vivo tumor killing activity of two OVs, vesicular stomatitis virus (VSV) and vaccinia virus is caused by indirect killing of uninfected tumor cells. Shortly after administering the oncolytic virus we observed limited virus infection, coincident with a loss of blood flow to the interior of the tumor that correlated with induction of apoptosis in tumor cells. Transcript profiling of tumors showed that virus infection resulted in a dramatic transcriptional activation of pro-inflammatory genes including the neutrophil chemoattractants CXCL1 and CXCL5. Immunohistochemical examination of infected tumors revealed infiltration by neutrophils correlating with chemokine induction. Depletion of neutrophils in animals prior to VSV administration eliminated uninfected tumor cell apoptosis and permitted more extensive replication and spreading of the virus throughout the tumor. Taken all together, these results indicate that targeted recruitment of neutrophils to infected tumor beds enhances the killing of malignant cells. We propose that activation of inflammatory cells can be used for enhancing the effectiveness of oncolytic virus therapeutics, and that this approach should influence the planning of therapeutic doses.
Collapse
Affiliation(s)
- Caroline J Breitbach
- Centre for Cancer Therapeutics, Ottawa Health Research Institute, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bluff JE, O'Ceallaigh S, O'Kane S, Ferguson MWJ, Ireland G. The microcirculation in acute murine cutaneous incisional wounds shows a spatial and temporal variation in the functionality of vessels. Wound Repair Regen 2007; 14:434-42. [PMID: 16939571 DOI: 10.1111/j.1743-6109.2006.00142.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A mouse perfusion model using fluorescently labeled dextran has been developed to investigate the functionality of blood vessels during cutaneous wound healing. By immunostaining cryostat sections of perfused wounds with antibodies that identify vessels, we were able to assess their functionality. There was an increase in the proportion of CD31(+)-perfused vessels in all wound regions with time, although the vessels of the wound margins and superficial granulation tissue (GT) took the longest to become perfused. More than 50% of the latter vessels were not perfused at 10 days postwounding. This is consistent with the growth of functional vessels from the wound base proceeding to the more superficial GT. The CD34 marker was expressed by a subpopulation of CD31(+) vessels. However, in contrast to CD31(+) vessels, the functionality of CD34(+) vessels did not change significantly with time and 50-75% of CD34(+) vessels in the GT and wound margins were nonfunctional. This might be explained either by apoptosis of the CD34(+) vessels or the loss of the marker with time. This study has important implications for assays of wound-healing angiogenesis based on histology and immunohistochemical markers for vessels, because vessel functionality differs both spatially and temporally during wound healing.
Collapse
Affiliation(s)
- Joanne E Bluff
- UK Centre for Tissue Engineering, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
20
|
Huang NF, Yu J, Sievers R, Li S, Lee RJ. Injectable biopolymers enhance angiogenesis after myocardial infarction. ACTA ACUST UNITED AC 2006; 11:1860-6. [PMID: 16411832 DOI: 10.1089/ten.2005.11.1860] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Novel strategies by which to repair ischemic myocardium after myocardial infarction include the use of three-dimensional polymer scaffolds. A comparative study was carried out to assess the therapeutic potential of fibrin, collagen I, and Matrigel as injectable biopolymers for repair after myocardial infarction. Using a rat model of left coronary artery occlusion followed by reperfusion, local injection of the biopolymers into the infarct zone yielded significantly higher levels of capillary formation, when compared with the saline control group, at 5 weeks posttreatment. However, the degree of angiogenesis was not significantly different among the biopolymers. In addition, the collagen biopolymer significantly enhanced infiltration of myofibroblasts into the infarct area when compared with the control group. The results of this study highlight the potential clinical benefit of these biopolymers as injectable scaffolds or cell delivery vehicles to the infarct zone after infarction.
Collapse
Affiliation(s)
- Ngan F Huang
- UCSF/UCB Joint Bioengineering Graduate Group, University of California Berkeley, Berkeley, California, USA
| | | | | | | | | |
Collapse
|
21
|
Christman KL, Fang Q, Kim AJ, Sievers RE, Fok HH, Candia AF, Colley KJ, Herradon G, Ezquerra L, Deuel TF, Lee RJ. Pleiotrophin induces formation of functional neovasculature in vivo. Biochem Biophys Res Commun 2005; 332:1146-52. [PMID: 15949466 DOI: 10.1016/j.bbrc.2005.04.174] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2005] [Accepted: 04/26/2005] [Indexed: 02/04/2023]
Abstract
Pleiotrophin (PTN) is a heparin-binding growth/differentiation inducing cytokine that shares 50% amino acid sequence identity and striking domain homology with Midkine (MK), the only other member of the Ptn/Mk developmental gene family. The Ptn gene is expressed in sites of early vascular development in embryos and in healing wounds and its constitutive expression in many human tumors is associated with an angiogenic phenotype, suggesting that PTN has an important role in angiogenesis during development and in wound repair and advanced malignancies. To directly test whether PTN is angiogenic in vivo, we injected a plasmid to express PTN into ischemic myocardium in rats. Pleiotrophin stimulated statistically significant increases in both normal appearing new capillaries and arterioles each of which had readily detectable levels of the arteriole marker, smooth muscle cell alpha-actin. Furthermore, the newly formed blood vessels were shown to interconnect with the existent coronary vascular system. The results of these studies demonstrate directly that PTN is an effective angiogenic agent in vivo able to initiate new vessel formation that is both normal in appearance and function. The data suggest that PTN signals the more "complete" new blood vessel formation through its ability to stimulate different functions in different cell types not limited to the endothelial cell.
Collapse
Affiliation(s)
- Karen L Christman
- University of California Berkeley and San Francisco Joint Bioengineering Graduate Group, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Luginbuehl V, Wenk E, Koch A, Gander B, Merkle HP, Meinel L. Insulin-like growth factor I-releasing alginate-tricalciumphosphate composites for bone regeneration. Pharm Res 2005; 22:940-50. [PMID: 15948038 DOI: 10.1007/s11095-005-4589-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Accepted: 02/22/2005] [Indexed: 12/13/2022]
Abstract
PURPOSE Development and characterization of an in situ-forming, osteoconductive, and growth factor-releasing bone implant. METHODS Injectable in situ-forming scaffolds were prepared from a 2% (m/v) alginate solution, tricalciumphosphate (TCP) granules, and poly(lactide-co-glycolide) microspheres (MS), loaded with the osteoinductive growth factor insulin-like growth factor I (IGF-I). Scaffolds were prepared by mixing the components followed by hydrogel formation through calcium carbonate-induced physical cross-linking of the alginate at slightly acidic pH. Physical-chemical properties and cell biocompatibility using osteoblast-like cells (MG-63 and Saos-2) of these scaffolds were investigated. RESULTS The addition of TCP to the alginate resulted in reduced swelling and gelation time and an increase in stiffness. Osteoblast-like cells (MG-63 and Saos-2) did not show toxic reactions and adhered circumferentially to the TCP granules surface. The addition of the IGF-I MS resulted in an up to sevenfold increased proliferation rate of MG-63 cells as compared to scaffold preparations without IGF-I MS. The alkaline phosphate (ALP) activity-a parameter for osteblastic activity-increased with increasing amounts of TCP in Saos-2 loaded composite scaffolds. CONCLUSIONS A prototype in situ-hardening composite system for conformal filling of bone defects supporting osteoblastic activity for further clinical testing in relevant fracture models was developed and characterized.
Collapse
Affiliation(s)
- Vera Luginbuehl
- Institute of Pharmaceutical Sciences, Drug Formulation and Delivery Group, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
23
|
Springer ML, Sievers RE, Viswanathan MN, Yee MS, Foster E, Grossman W, Yeghiazarians Y. Closed-chest cell injections into mouse myocardium guided by high-resolution echocardiography. Am J Physiol Heart Circ Physiol 2005; 289:H1307-14. [PMID: 15908468 DOI: 10.1152/ajpheart.00164.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mouse is an important model for the development of therapeutic stem cell/bone marrow cell implantation to treat ischemic myocardium. However, its small heart size hampers accurate implantation into the left ventricular (LV) wall. Precise injections have required surgical visualization of the heart, which is subject to complications and is impractical for delayed or repeated injections. Furthermore, the thickness of the myocardium is comparable to the length of a needle bevel, so surgical exposure does not prevent inadvertent injection into the LV cavity. We describe the use of high-resolution echocardiography to guide nonsurgical injections accurately into the mouse myocardial wall. We optimized this system by using a mixture of ultrasound contrast and fluorescent microspheres injected into the myocardium, which enabled us to interpret the ultrasound image of the needle during injection. Quantitative dye injection studies demonstrated that guided closed-chest injections and open-chest injections deliver comparable amounts of injectate to the myocardium. We successfully used this system in a mouse myocardial infarction model to target the injection of labeled cells to a region adjacent to the infarct. Intentional injection of tracer into the LV cavity resulted in a small accumulation in the myocardium, suggesting that non-guided cell injections into mouse hearts may appear to be successful even if the majority of the injectate is lost in the chamber. The use of this system will allow more precise cellular implantation into the mouse myocardium by accurately guiding injections to desired locations, confirming successful implantation of cells, in a clinically relevant time frame.
Collapse
Affiliation(s)
- Matthew L Springer
- Division of Cardiology, Department of Medicine, University of California, San Francisco, California 94143-0124, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Christman KL, Fang Q, Yee MS, Johnson KR, Sievers RE, Lee RJ. Enhanced neovasculature formation in ischemic myocardium following delivery of pleiotrophin plasmid in a biopolymer. Biomaterials 2005; 26:1139-44. [PMID: 15451633 DOI: 10.1016/j.biomaterials.2004.04.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 04/08/2004] [Indexed: 11/18/2022]
Abstract
Coronary heart disease is currently the leading killer in the western world. Therapeutic angiogenic agents are currently being examined for treatment of this disease. We have recently demonstrated the effective use of Pleiotrophin (PTN) as a therapeutic agent for treatment of ischemic myocardium. We have also shown that injection of the biopolymer fibrin glue preserves left ventricular geometry and prevents a deterioration of cardiac function following myocardial infarction. Due to the low transfection efficiency of naked plasmid injections, we examined the use of PTN plasmid and the biopolymer as a gene-activated matrix in the myocardium. In this study, we demonstrate that delivery of PTN plasmid in fibrin glue increases neovasculature formation compared to injection of the naked plasmid in saline.
Collapse
Affiliation(s)
- Karen L Christman
- University of California Berkeley and San Francisco Joint Bioengineering Graduate Group, San Francisco, CA 94143, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Mitra S, Maugain E, Bolotine L, Guillemin F, Foster TH. Temporally and Spatially Heterogeneous Distribution of mTHPC in a Murine Tumor Observed by Two-color Confocal Fluorescence Imaging and Spectroscopy in a Whole-mount Model. Photochem Photobiol 2005; 81:1123-30. [PMID: 15971933 DOI: 10.1562/2005-03-24-ra-471] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Efficient intratumor delivery of anticancer drugs and photosensitizers is an important factor in the success of chemotherapy and photodynamic therapy, respectively. Unfortunately, their adequate and uniform intratumor distribution is impeded by several physiological barriers and by binding to tissue components. Measurement of gross tumor drug accumulation is a routine method of investigating the uptake and clearance of chemotherapy agents and photosensitizers but tells little about their extravascular spatial distribution. We use whole-mount two-color confocal fluorescence imaging and imaging spectroscopy of unprocessed excised murine tumor fragments to investigate the intratumor distribution of the photosensitizer meso-tetrahydroxyphenyl chlorin (mTHPC) as a function of distance from blood vessels perfused with 0.2 mum diameter fluorescent microspheres. Significant mismatches between drug and perfused vasculature are caused by heterogeneities in tumor blood supply. We describe complex microscopic mTHPC gradients that reverse dramatically relative to the perfused vasculature with time after injection. This imaging technique can be applied to screen the dynamic intratumor distribution of other fluorescent photosensitizers and anticancer drugs.
Collapse
Affiliation(s)
- Soumya Mitra
- Department of Radiology, University of Rochester, Rochester, NY, USA
| | | | | | | | | |
Collapse
|
26
|
Gu F, Amsden B, Neufeld R. Sustained delivery of vascular endothelial growth factor with alginate beads. J Control Release 2004; 96:463-72. [PMID: 15120902 DOI: 10.1016/j.jconrel.2004.02.021] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2003] [Accepted: 02/26/2004] [Indexed: 10/26/2022]
Abstract
Therapeutic angiogenesis is the growth of blood vessels from a pre-existing vasculature for clinical applications such as treating myocardial and limb ischemia. Vascular endothelial growth factor (VEGF) is a potent signal transduction molecule that acts specifically on vascular endothelial cells. Encapsulation of VEGF in a polymer matrix not only protects protein against enzymatic degradation in the body, but also allows proteins to be released at a controllable rate into a localized area. In this study, VEGF was encapsulated in calcium alginate beads by the extrusion/external gelation method, and was subsequently released in PBS and in serum media. The objective was to optimize VEGF encapsulation yield and obtain VEGF release at a constant rate from alginate matrices in vitro. The incorporation of low concentrations of VEGF and NaCl can increase encapsulation yield to 97%. The rate of VEGF release from alginate beads was higher in serum than in PBS, which was due to the capacity of the serum in reducing the electrostatic interaction between alginate and VEGF. The presence of CaCl(2) in the release supernatant can shield the alginate interaction with VEGF, and a constant release rate of 6 ng/ml/day may be sustained for 14 days. These results suggest that the alginate-VEGF delivery system may be useful in the development of vascular tissue engineering and wound healing applications.
Collapse
Affiliation(s)
- Frank Gu
- Department of Chemical Engineering, Queen's University, Kingston, ON, Canada K7L 3N6
| | | | | |
Collapse
|
27
|
Brühl T, Heeschen C, Aicher A, Jadidi AS, Haendeler J, Hoffmann J, Schneider MD, Zeiher AM, Dimmeler S, Rössig L. p21Cip1 levels differentially regulate turnover of mature endothelial cells, endothelial progenitor cells, and in vivo neovascularization. Circ Res 2004; 94:686-92. [PMID: 14752032 DOI: 10.1161/01.res.0000119922.71855.56] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
p21(Cip1) (p21) controls cell cycle progression and apoptosis in mature endothelial cells (ECs) and regulates size and cycling of the hematopoietic progenitor cell pool. Because circulating endothelial progenitor cells (EPCs) contribute to postnatal neovascularization in addition to mature ECs, we investigated the regulation of ECs and EPCs in p21-deficient mice. Mature aortic EC proliferation was increased in homozygous p21(-/-) and heterozygous p21(+/-) mice, in which p21 protein levels are reduced to one third of wild-type (WT). In contrast, apoptosis sensitivity was increased by 3.5-fold only in p21(-/-), but not in p21(+/-) mice. Consistently, in vivo apoptosis of ECs within areas of neovascularization was elevated in p21(-/-) but not in p21(+/-) mice. EPC numbers were elevated 2-fold in p21(-/-) mice compared with WT (P<0.001), and clonal expansion capacity of EPCs was increased from 25+/-4 (WT) to 57+/-8 colony-forming units in p21(-/-) mice (P<0.005). EPC numbers and expansion were likewise increased in p21(+/-) mice. As the integrative endpoint, in vivo neovascularization reflecting all p21-affected parameters was increased over WT only in p21(+/-) (P<0.001), but not in p21(-/-) mice. In conclusion, reduced p21 protein levels of mice lacking one p21 allele are associated with increased proliferation of ECs and EPCs, whereas survival of ECs to apoptotic stimuli in vitro and in vivo is not impaired. Under these conditions, neovascularization was increased. In contrast, complete p21 deficiency did not result in an increased neovascularization despite increased mature EC and EPC proliferation. This may be due to the sensitization of ECs against apoptosis.
Collapse
Affiliation(s)
- Tom Brühl
- Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sheng Y, Hua J, Pinney KG, Garner CM, Kane RR, Prezioso JA, Chaplin DJ, Edvardsen K. Combretastatin family member OXI4503 induces tumor vascular collapse through the induction of endothelial apoptosis. Int J Cancer 2004; 111:604-10. [PMID: 15239140 DOI: 10.1002/ijc.20297] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mechanism of tumor cell killing by OXI4503 was investigated by studying vascular functional and morphological changes post drug administration. SCID mice bearing MHEC5-T hemangioendothelioma were given a single dose of OXI4503 at 100 mg/kg. Tumor blood flow, measured by microsphere fluorescence, was reduced by 50% at 1 hr, and reached a maximum level 6-24 hr post drug treatment. Tumor vascular permeability, measured by Evan's blue and hemoglobin, increased significantly from 3 hr and peaked at 18 hr. The elevated tumor vessel permeability was accompanied by an increase in vascular endothelial growth factor (VEGF) from 1 hr post drug treatment. Immunohistochemical staining for CD31 and laminin showed that tumor blood vessels were affected as early as 3 hr but more prominent from 6 hr. From 12 hr, the vessel structure was completely destroyed. Histopathological and double immunohistochemical staining showed morphological change and induction of apoptosis in endothelial cells at 1-3 hr, followed by tumor cell necrosis from 6-72 hr. There were no statistically significant changes of Evan's blue and hemoglobin contents in liver tissue over the time course. These results suggest that OXI4503 selectively targets tumor blood vessels, and induces blood flow shutdown while it enhances tumor blood vessel permeability. The early induction of endothelial cell apoptosis leads to functional changes of tumor blood vessels and finally to the collapse of tumor vasculature, resulting in massive tumor cell necrosis. The time course of the tumor vascular response observed with OXI4503 treatment supports this drug for development as a stand alone therapy, and also lends support for the use of the drug in combination with other cancer therapies.
Collapse
Affiliation(s)
- Yezhou Sheng
- Department of Cell and Molecular Biology, Section for Tumor Immunology, University of Lund, Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood 2003; 102:1340-6. [PMID: 12702503 DOI: 10.1182/blood-2003-01-0223] [Citation(s) in RCA: 667] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence suggests that postnatal neovascularization involves the recruitment of circulating endothelial progenitor cells (EPCs). Hematopoietic and endothelial cell lineages share common progenitors. Cytokines formerly thought to be specific for the hematopoietic system have only recently been shown to affect several functions in endothelial cells. Accordingly, we investigated the stimulatory potential of erythropoietin (Epo) on EPC mobilization and neovascularization. The bone marrow of Epo-treated mice showed a significant increase in number and proliferation of stem and progenitor cells as well as in colony-forming units. The number of isolated EPCs and CD34+/flk-1+ precursor cells was significantly increased in spleen and peripheral blood of Epo-treated mice compared with phosphate-buffered saline-treated mice. In in vivo models of postnatal neovascularization, Epo significantly increased inflammation- and ischemia-induced neovascularization. The physiologic relevance of these findings was investigated in patients with coronary heart disease. In a multivariate regression model, serum levels of Epo and vascular endothelial growth factor were significantly associated with the number of stem and progenitor cells in the bone marrow as well as with the number and function of circulating EPCs. In conclusion, the present study suggests that Epo stimulates postnatal neovascularization at least in part by enhancing EPC mobilization from the bone marrow. Epo appears to physiologically regulate EPC mobilization in patients with ischemic heart disease. Thus, Epo serum levels may help in identifying patients with impaired EPC recruitment capacity.
Collapse
Affiliation(s)
- Christopher Heeschen
- Department of Internal Medicine IV and Hematology, University of Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Springer ML, Ozawa CR, Banfi A, Kraft PE, Ip TK, Brazelton TR, Blau HM. Localized arteriole formation directly adjacent to the site of VEGF-induced angiogenesis in muscle. Mol Ther 2003; 7:441-9. [PMID: 12727106 DOI: 10.1016/s1525-0016(03)00010-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We have shown previously that implantation of myoblasts constitutively expressing the VEGF-A gene into nonischemic mouse skeletal muscle leads to overgrowth of capillary-like blood vessels and hemangioma formation. These aberrant effects occurred directly at the implantation site. We show here that these regions result from angiogenic capillary growth and involve a change in capillary growth pattern and that smooth muscle-coated vessels similar to arterioles form directly adjacent to the implantation site. Myoblasts genetically engineered to produce VEGF were implanted into mouse leg muscles. Implantation sites were surrounded by a zone of dense capillary-sized vessels, around which was a second zone of muscle containing larger, smooth-muscle-covered vessels but few capillaries, and an outer zone of muscle exhibiting normal capillary density. The lack of capillaries in the middle region suggests that the preexisting capillaries adjacent to the implantation site underwent enlargement and/or fusion and recruited a smooth muscle coat. Capillaries at the implantation site were frequently wrapped around VEGF-producing muscle fibers and were continuous with the circulation and were not observed to include bone-marrow-derived endothelial cells. In contrast with the distant arteriogenesis resulting from VEGF delivery described in previous studies, we report here that highly localized arterioles also form adjacent to the site of delivery.
Collapse
Affiliation(s)
- Matthew L Springer
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, California 94305-5175, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Spurbeck WW, Ng CYC, Strom TS, Vanin EF, Davidoff AM. Enforced expression of tissue inhibitor of matrix metalloproteinase-3 affects functional capillary morphogenesis and inhibits tumor growth in a murine tumor model. Blood 2002; 100:3361-8. [PMID: 12384438 DOI: 10.1182/blood.v100.9.3361] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Homeostasis of the extracellular matrix is a delicate balance between degradation and remodeling, the balance being maintained by the interaction of activated matrix metalloproteinases (MMPs) and specific tissue inhibitors of matrix metalloproteinases (TIMPs). Up-regulation of MMP activity, favoring proteolytic degradation of the basement membrane and extracellular matrix, has been linked to tumor growth and metastasis, as well as tumor-associated angiogenesis, whereas inhibition of MMP activity appears to restrict these processes. We have used retroviral-mediated gene delivery to effect sustained autocrine expression of TIMP-3 in murine neuroblastoma and melanoma tumor cells in order to further examine the ability of TIMPs to inhibit angiogenesis in vivo. Growth of both histologic types of gene-modified tumor cells in severe combined immunodeficiency (SCID) mice was significantly restricted when compared with controls. Grossly, these tumors were small and had few feeding vessels. Histologic evaluation revealed that although tumors overexpressing TIMP-3 had an increased number of CD31(+) endothelial cells, these endothelial cells had not formed functional tubules, as evidenced by decreased vessel continuity and minimal pericyte recruitment. This effect appears to be mediated, in part, by decreased expression of vascular endothelial (VE)-cadherin by endothelial cells in the presence of TIMP-3 as seen both in an in vitro assay and in TIMP-3-overexpressing tumors. Taken together, these results demonstrate that overexpression of TIMP-3 can inhibit angiogenesis and associated tumor growth, and that the antiangiogenic effects of TIMP-3 appear to be mediated through the inhibition of functional capillary morphogenesis.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Autocrine Communication
- Cadherins/biosynthesis
- Capillaries/ultrastructure
- Cell Hypoxia
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/pathology
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Green Fluorescent Proteins
- Luminescent Proteins/analysis
- Luminescent Proteins/genetics
- Matrix Metalloproteinases/physiology
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, SCID
- Microcirculation
- Morphogenesis
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neovascularization, Pathologic/metabolism
- Neuroblastoma/blood supply
- Neuroblastoma/metabolism
- Neuroblastoma/pathology
- Platelet Endothelial Cell Adhesion Molecule-1/analysis
- Recombinant Fusion Proteins/physiology
- Tissue Inhibitor of Metalloproteinase-3/biosynthesis
- Tissue Inhibitor of Metalloproteinase-3/genetics
- Tissue Inhibitor of Metalloproteinase-3/physiology
- Transfection
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
Collapse
Affiliation(s)
- William W Spurbeck
- Departments of Surgery and Hematology/Oncology, St Jude Children's Research Hospital, 332 S. Lauderdale, Memphis, TN 38105, USA
| | | | | | | | | |
Collapse
|
32
|
Heeschen C, Weis M, Aicher A, Dimmeler S, Cooke JP. A novel angiogenic pathway mediated by non-neuronal nicotinic acetylcholine receptors. J Clin Invest 2002. [DOI: 10.1172/jci0214676] [Citation(s) in RCA: 225] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
33
|
Heeschen C, Weis M, Aicher A, Dimmeler S, Cooke JP. A novel angiogenic pathway mediated by non-neuronal nicotinic acetylcholine receptors. J Clin Invest 2002; 110:527-36. [PMID: 12189247 PMCID: PMC150415 DOI: 10.1172/jci14676] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We have recently reported that nicotine has angiogenic effects, which appear to be mediated through non-neuronal nicotinic acetylcholine receptors (nAChRs). Here, we describe the endogenous cholinergic pathway for angiogenesis. In an in vitro angiogenesis model, increasing concentrations of the nonselective nAChR antagonist mecamylamine completely and reversibly inhibited endothelial network formation. Although several nAChR isoforms are expressed on endothelial cells (ECs), a similar inhibition was only obtained with the selective alpha7-nAChR antagonist alpha-bungarotoxin, whereas other selective antagonists did not result in significant inhibition of network formation. alpha7-nAChR was upregulated during proliferation, by hypoxia in vitro, and by ischemia in vivo. The nAChR-induced network formation was partially dependent on VEGF, was completely dependent on the phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathways, and finally resulted in NF-kappaB activation. In vivo, pharmacological inhibition of nAChR as well as genetic disruption of alpha7-nAChR expression significantly inhibited inflammatory angiogenesis and reduced ischemia-induced angiogenesis and tumor growth. Our results suggest that nAChRs may play an important role in physiological and pathological angiogenesis. To our knowledge, this is the first description of a cholinergic angiogenic pathway, and it suggests a novel avenue for therapeutic modulation of angiogenesis.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/pathology
- Cell Division/drug effects
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/metabolism
- Hindlimb/blood supply
- Humans
- Ischemia/pathology
- Mecamylamine/pharmacology
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Nicotinic Antagonists/pharmacology
- Receptors, Nicotinic/metabolism
- Receptors, Nicotinic/physiology
- alpha7 Nicotinic Acetylcholine Receptor
Collapse
Affiliation(s)
- Christopher Heeschen
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | |
Collapse
|
34
|
Affiliation(s)
- Andrea Banfi
- Department of Molecular Pharmacology, Stanford University School of Medicine, CCSR 4215, Stanford, California 94305, USA
| | | | | |
Collapse
|
35
|
Abstract
BACKGROUND Statins inhibit HMG-CoA reductase to reduce the synthesis of cholesterol and isoprenoids that modulate diverse cell functions. We investigated the effect of the statins cerivastatin and atorvastatin on angiogenesis in vitro and in vivo. METHODS AND RESULTS Endothelial cell proliferation, migration, and differentiation were enhanced at low concentrations (0.005 to 0.01 micromol/L) but significantly inhibited at high statin concentrations (0.05 to 1 micromol/L). Antiangiogenic effects at high concentrations were associated with decreased endothelial release of vascular endothelial growth factor and increased endothelial apoptosis and were reversed by geranylgeranyl pyrophosphate. In murine models, inflammation-induced angiogenesis was enhanced with low-dose statin therapy (0.5 mg x kg(-1) x d(-1)) but significantly inhibited with high concentrations of cerivastatin or atorvastatin (2.5 mg x kg(-1) x d(-1)). Despite the fact that high-dose statin treatment was effective at reducing lipid levels in hyperlipidemic apolipoprotein E-deficient mice, it impaired rather than enhanced angiogenesis. Finally, high-dose cerivastatin decreased tumor growth and tumor vascularization in a murine Lewis lung cancer model. CONCLUSIONS HMG-CoA reductase inhibition has a biphasic dose-dependent effect on angiogenesis that is lipid independent and associated with alterations in endothelial apoptosis and vascular endothelial growth factor signaling. Statins have proangiogenic effects at low therapeutic concentrations but angiostatic effects at high concentrations that are reversed by geranylgeranyl pyrophosphate. At clinically relevant doses, statins may modulate angiogenesis in humans via effects on geranylated proteins.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Apoptosis/drug effects
- Atorvastatin
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/pathology
- Cell Differentiation/drug effects
- Cell Division/drug effects
- Cell Movement/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Endothelial Growth Factors/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Female
- Heptanoic Acids/pharmacology
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Hyperlipidemias/drug therapy
- Hyperlipidemias/genetics
- Lymphokines/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Neovascularization, Physiologic/drug effects
- Polyisoprenyl Phosphates/pharmacology
- Pyridines/pharmacology
- Pyrroles/pharmacology
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptors, Growth Factor/biosynthesis
- Receptors, Vascular Endothelial Growth Factor
- Signal Transduction/drug effects
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- Michael Weis
- Stanford University School of Medicine, Division of Cardiovascular Medicine, Stanford, Calif 94305, USA
| | | | | | | |
Collapse
|