101
|
Shay JES, Imtiyaz HZ, Sivanand S, Durham AC, Skuli N, Hsu S, Mucaj V, Eisinger-Mathason TSK, Krock BL, Giannoukos DN, Simon MC. Inhibition of hypoxia-inducible factors limits tumor progression in a mouse model of colorectal cancer. Carcinogenesis 2014; 35:1067-77. [PMID: 24408928 DOI: 10.1093/carcin/bgu004] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hypoxia-inducible factors (HIFs) accumulate in both neoplastic and inflammatory cells within the tumor microenvironment and impact the progression of a variety of diseases, including colorectal cancer. Pharmacological HIF inhibition represents a novel therapeutic strategy for cancer treatment. We show here that acriflavine (ACF), a naturally occurring compound known to repress HIF transcriptional activity, halts the progression of an autochthonous model of established colitis-associated colon cancer (CAC) in immunocompetent mice. ACF treatment resulted in decreased tumor number, size and advancement (based on histopathological scoring) of CAC. Moreover, ACF treatment corresponded with decreased macrophage infiltration and vascularity in colorectal tumors. Importantly, ACF treatment inhibited the hypoxic induction of M-CSFR, as well as the expression of the angiogenic factor (vascular endothelial growth factor), a canonical HIF target, with little to no impact on the Nuclear factor-kappa B pathway in bone marrow-derived macrophages. These effects probably explain the observed in vivo phenotypes. Finally, an allograft tumor model further confirmed that ACF treatment inhibits tumor growth through HIF-dependent mechanisms. These results suggest pharmacological HIF inhibition in multiple cell types, including epithelial and innate immune cells, significantly limits tumor growth and progression.
Collapse
|
102
|
Baginska J, Viry E, Paggetti J, Medves S, Berchem G, Moussay E, Janji B. The critical role of the tumor microenvironment in shaping natural killer cell-mediated anti-tumor immunity. Front Immunol 2013; 4:490. [PMID: 24400010 PMCID: PMC3872331 DOI: 10.3389/fimmu.2013.00490] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/13/2013] [Indexed: 12/27/2022] Open
Abstract
Considerable evidence has been gathered over the last 10 years showing that the tumor microenvironment (TME) is not simply a passive recipient of immune cells, but an active participant in the establishment of immunosuppressive conditions. It is now well documented that hypoxia, within the TME, affects the functions of immune effectors including natural killer (NK) cells by multiple overlapping mechanisms. Indeed, each cell in the TME, irrespective of its transformation status, has the capacity to adapt to the hostile TME and produce immune modulatory signals or mediators affecting the function of immune cells either directly or through the stimulation of other cells present in the tumor site. This observation has led to intense research efforts focused mainly on tumor-derived factors. Notably, it has become increasingly clear that tumor cells secrete a number of environmental factors such as cytokines, growth factors, exosomes, and microRNAs impacting the immune cell response. Moreover, tumor cells in hostile microenvironments may activate their own intrinsic resistance mechanisms, such as autophagy, to escape the effective immune response. Such adaptive mechanisms may also include the ability of tumor cells to modify their metabolism and release several metabolites to impair the function of immune cells. In this review, we summarize the different mechanisms involved in the TME that affect the anti-tumor immune function of NK cells.
Collapse
Affiliation(s)
- Joanna Baginska
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| | - Elodie Viry
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| | - Jérôme Paggetti
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| | - Sandrine Medves
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| | - Guy Berchem
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| | - Etienne Moussay
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| | - Bassam Janji
- Laboratory of Experimental Hemato-Oncology, Department of Oncology, Public Research Center for Health (CRP-Santé) , Luxembourg City , Luxembourg
| |
Collapse
|
103
|
Casazza A, Laoui D, Wenes M, Rizzolio S, Bassani N, Mambretti M, Deschoemaeker S, Van Ginderachter JA, Tamagnone L, Mazzone M. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 2013; 24:695-709. [PMID: 24332039 DOI: 10.1016/j.ccr.2013.11.007] [Citation(s) in RCA: 477] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 10/04/2013] [Accepted: 11/10/2013] [Indexed: 10/25/2022]
Abstract
Recruitment of tumor-associated macrophages (TAMs) into avascular areas sustains tumor progression; however, the underlying guidance mechanisms are unknown. Here, we report that hypoxia-induced Semaphorin 3A (Sema3A) acts as an attractant for TAMs by triggering vascular endothelial growth factor receptor 1 phosphorylation through the associated holoreceptor, composed of Neuropilin-1 (Nrp1) and PlexinA1/PlexinA4. Importantly, whereas Nrp1 levels are downregulated in the hypoxic environment, Sema3A continues to regulate TAMs in an Nrp1-independent manner by eliciting PlexinA1/PlexinA4-mediated stop signals, which retain them inside the hypoxic niche. Consistently, gene deletion of Nrp1 in macrophages favors TAMs' entrapment in normoxic tumor regions, which abates their pro-angiogenic and immunosuppressive functions, hence inhibiting tumor growth and metastasis. This study shows that TAMs' heterogeneity depends on their localization, which is tightly controlled by Sema3A/Nrp1 signaling.
Collapse
Affiliation(s)
- Andrea Casazza
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Damya Laoui
- Laboratory of Myeloid Cell Immunology, VIB, 1050 Brussels, Belgium; Laboratory of Cellular and Molecular Immunology, Department of Molecular and Cellular Interactions, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Mathias Wenes
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Sabrina Rizzolio
- Institute for Cancer Research at Candiolo, Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
| | - Nicklas Bassani
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Marco Mambretti
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Sofie Deschoemaeker
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Jo A Van Ginderachter
- Laboratory of Myeloid Cell Immunology, VIB, 1050 Brussels, Belgium; Laboratory of Cellular and Molecular Immunology, Department of Molecular and Cellular Interactions, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Luca Tamagnone
- Institute for Cancer Research at Candiolo, Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
| | - Massimiliano Mazzone
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, 3000 Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Department of Oncology, Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
104
|
Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, Movahedi K, Houbracken I, Schouppe E, Elkrim Y, Karroum O, Jordan B, Carmeliet P, Gysemans C, De Baetselier P, Mazzone M, Van Ginderachter JA. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res 2013; 74:24-30. [PMID: 24220244 DOI: 10.1158/0008-5472.can-13-1196] [Citation(s) in RCA: 311] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor-associated macrophages (TAM) are exposed to multiple microenvironmental cues in tumors, which collaborate to endow these cells with protumoral activities. Hypoxia, caused by an imbalance in oxygen supply and demand because of a poorly organized vasculature, is often a prominent feature in solid tumors. However, to what extent tumor hypoxia regulates the TAM phenotype in vivo is unknown. Here, we show that the myeloid infiltrate in mouse lung carcinoma tumors encompasses two morphologically distinct CD11b(hi)F4/80(hi)Ly6C(lo) TAM subsets, designated as MHC-II(lo) and MHC-II(hi) TAM, both of which were derived from tumor-infiltrating Ly6C(hi) monocytes. MHC-II(lo) TAM express higher levels of prototypical M2 markers and reside in more hypoxic regions. Consequently, MHC-II(lo) TAM contain higher mRNA levels for hypoxia-regulated genes than their MHC-II(hi) counterparts. To assess the in vivo role of hypoxia on these TAM features, cancer cells were inoculated in prolyl hydroxylase domain 2 (PHD2)-haplodeficient mice, resulting in better-oxygenated tumors. Interestingly, reduced tumor hypoxia did not alter the relative abundance of TAM subsets nor their M2 marker expression, but specifically lowered hypoxia-sensitive gene expression and angiogenic activity in the MHC-II(lo) TAM subset. The same observation in PHD2(+/+) → PHD2(+/-) bone marrow chimeras also suggests organization of a better-oxygenized microenvironment. Together, our results show that hypoxia is not a major driver of TAM subset differentiation, but rather specifically fine-tunes the phenotype of M2-like MHC-II(lo) TAM.
Collapse
Affiliation(s)
- Damya Laoui
- Authors' Affiliations: Laboratory of Myeloid Cell Immunology, VIB; Laboratory of Cellular and Molecular Immunology; Cell Differentiation Unit, Diabetes Research Centre, Vrije Universiteit Brussel; Biomedical Magnetic Resonance Unit, U.C. Louvain, Brussels; Laboratory of Molecular Oncology and Angiogenesis; Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB; and Experimental Medicine and Endocrinology, Department of Experimental Medicine, K.U. Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Strehl C, Fangradt M, Fearon U, Gaber T, Buttgereit F, Veale DJ. Hypoxia: how does the monocyte-macrophage system respond to changes in oxygen availability? J Leukoc Biol 2013; 95:233-41. [PMID: 24168857 DOI: 10.1189/jlb.1212627] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hypoxia is an important feature of inflamed tissue, such as the RA joint. Activated monocytes/macrophages and endothelial cells play a pivotal role in the pathogenesis of RA, implicated in the mechanism of inflammation and erosion. During development, myeloid progenitor cells sequentially give rise to monoblasts, promonocytes, and monocytes that are released from the bone marrow into the bloodstream. After extravasation, monocytes differentiate into long-lived, tissue-specific macrophages or DCs. The effect of different oxygen concentrations experienced by these cells during maturation represents a novel aspect of this developmental process. In inflamed joint tissue, the microvascular architecture is highly dysregulated; thus, efficiency of oxygen supply to the synovium is poor. Therefore, invading cells must adapt instantaneously to changes in the oxygen level of the microenvironment. Angiogenesis is an early event in the inflammatory joint, which is important in enabling activated monocytes to enter via endothelial cells by active recruitment to expand the synovium into a "pannus", resulting in cartilage degradation and bone destruction. The increased metabolic turnover of the expanding synovial pannus outpaces the dysfunctional vascular supply, resulting in hypoxia. The abnormal bioenergetics of the microenvironment further promotes synovial cell invasiveness. In RA, joint hypoxia represents a potential threat to cell function and survival. Notably, oxygen availability is a crucial parameter in the cellular energy metabolism, itself an important factor in determining the function of immune cells.
Collapse
Affiliation(s)
- Cindy Strehl
- 2.Charité-Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
106
|
Oxidative stress induced mitochondrial protein kinase A mediates cytochrome c oxidase dysfunction. PLoS One 2013; 8:e77129. [PMID: 24130844 PMCID: PMC3795003 DOI: 10.1371/journal.pone.0077129] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/29/2013] [Indexed: 11/30/2022] Open
Abstract
Previously we showed that Protein kinase A (PKA) activated in hypoxia and myocardial ischemia/reperfusion mediates phosphorylation of subunits I, IVi1 and Vb of cytochrome c oxidase. However, the mechanism of activation of the kinase under hypoxia remains unclear. It is also unclear if hypoxic stress activated PKA is different from the cAMP dependent mitochondrial PKA activity reported under normal physiological conditions. In this study using RAW 264.7 macrophages and in vitro perfused mouse heart system we investigated the nature of PKA activated under hypoxia. Limited protease treatment and digitonin fractionation of intact mitochondria suggests that higher mitochondrial PKA activity under hypoxia is mainly due to increased sequestration of PKA Catalytic α (PKAα) subunit in the mitochondrial matrix compartment. The increase in PKA activity is independent of mitochondrial cAMP and is not inhibited by adenylate cyclase inhibitor, KH7. Instead, activation of hypoxia-induced PKA is dependent on reactive oxygen species (ROS). H89, an inhibitor of PKA activity and the antioxidant Mito-CP prevented loss of CcO activity in macrophages under hypoxia and in mouse heart under ischemia/reperfusion injury. Substitution of wild type subunit Vb of CcO with phosphorylation resistant S40A mutant subunit attenuated the loss of CcO activity and reduced ROS production. These results provide a compelling evidence for hypoxia induced phosphorylation as a signal for CcO dysfunction. The results also describe a novel mechanism of mitochondrial PKA activation which is independent of mitochondrial cAMP, but responsive to ROS.
Collapse
|
107
|
Hypoxia/ischemia up-regulates Id2 expression in neuronal cells in vivo and in vitro. Neurosci Lett 2013; 554:88-93. [PMID: 23994058 DOI: 10.1016/j.neulet.2013.08.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 11/21/2022]
Abstract
Inhibitor of DNA binding/differentiation 2 (Id2) belongs to a family of transcriptional modulators characterized by a helix-loop-helix (HLH) motif that lacks the basic amino acid domain necessary to bind DNA. The aim of this study was to obtain a better understanding of the role of Id2 in hypoxia/ischemia (H/I)-induced neuronal apoptosis. Following H/I induction in a rat model of middle cerebral artery occlusion (MCAO)/reperfusion, the number of TUNEL-positive cells in cerebral cortices of the penumbra area increased gradually, while the Id2 mRNA and protein expression were also significantly up-regulated. The hypoxia-mimetic, cobalt chloride (CoCl2)-treated rat neuroblastoma B35 cell line also demonstrated enhanced Id2 mRNA and protein expression as well as increased number of cells in the sub-G1 populations after H/I exposure. Consistently, the expression of Bax, a proapoptotic protein, was also up-regulated in vivo and in vitro. Moreover, triple immunofluorescence demonstrated the obvious co-localization of Id2, TUNEL and NeuN in neurons of the penumbra area. These data suggest that H/I up-regulates Id2 expression in neuronal cells, and Id2 might play an important role in H/I-induced neuronal apoptosis.
Collapse
|
108
|
Zhou J, Zhou S. Inflammation: therapeutic targets for diabetic neuropathy. Mol Neurobiol 2013; 49:536-46. [PMID: 23990376 DOI: 10.1007/s12035-013-8537-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/15/2013] [Indexed: 11/26/2022]
Abstract
There are still no approved treatments for the prevention or of cure of diabetic neuropathy, and only symptomatic pain therapies of variable efficacy are available. Inflammation is a cardinal pathogenic mechanism of diabetic neuropathy. The relationships between inflammation and the development of diabetic neuropathy involve complex molecular networks and processes. Herein, we review the key inflammatory molecules (inflammatory cytokines, adhesion molecules, chemokines) and pathways (nuclear factor kappa B, JUN N-terminal kinase) implicated in the development and progression of diabetic neuropathy. Advances in the understanding of the roles of these key inflammatory molecules and pathways in diabetic neuropathy will facilitate the discovery of the potential of anti-inflammatory approaches for the inhibition of the development of neuropathy. Specifically, many anti-inflammatory drugs significantly inhibit the development of different aspects of diabetic neuropathy in animal models and clinical trials.
Collapse
Affiliation(s)
- Jiyin Zhou
- National Drug Clinical Trial Institution, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China,
| | | |
Collapse
|
109
|
Russell JS, Brown JM. The irradiated tumor microenvironment: role of tumor-associated macrophages in vascular recovery. Front Physiol 2013; 4:157. [PMID: 23882218 PMCID: PMC3713331 DOI: 10.3389/fphys.2013.00157] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is an important modality used in the treatment of more than 50% of cancer patients in the US. However, despite sophisticated techniques for radiation delivery as well as the combination of radiation with chemotherapy, tumors can recur. Thus, any method of improving the local control of the primary tumor by radiotherapy would produce a major improvement in the curability of cancer patients. One of the challenges in the field is to understand how the tumor vasculature can regrow after radiation in order to support tumor recurrence, as it is unlikely that any of the endothelial cells within the tumor could survive the doses given in a typical radiotherapy regimen. There is now considerable evidence from both preclinical and clinical studies that the tumor vasculature can be restored following radiotherapy from an influx of circulating cells consisting primarily of bone marrow derived monocytes and macrophages. The radiation-induced influx of bone marrow derived cells (BMDCs) into tumors can be prevented through the blockade of various cytokine pathways and such strategies can inhibit tumor recurrence. However, the post-radiation interactions between surviving tumor cells, recruited immune cells, and the remaining stroma remain poorly defined. While prior studies have described the monocyte/macrophage inflammatory response within normal tissues and in the tumor microenvironment, less is known about this response with respect to a tumor after radiation therapy. The goal of this review is to summarize existing research studies to provide an understanding of how the myelomonocytic lineage may influence vascular recovery within the irradiated tumor microenvironment.
Collapse
Affiliation(s)
- Jeffery S Russell
- Department of Medical Oncology, Stanford University School of Medicine Stanford, CA, USA
| | | |
Collapse
|
110
|
Akash MSH, Rehman K, Chen S. Role of inflammatory mechanisms in pathogenesis of type 2 diabetes mellitus. J Cell Biochem 2013; 114:525-31. [PMID: 22991242 DOI: 10.1002/jcb.24402] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/11/2012] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by progressive β-cell dysfunctioning and insulin resistance. This article reviews recent literature with special focus on inflammatory mechanisms that provoke the pathogenesis of T2DM. We have focused on the recent advances in progression of T2DM including various inflammatory mechanisms that might induce inflammation, insulin resistance, decrease insulin secretion from pancreatic islets and dysfunctioning of β-cells. Here we have also summarized the role of various pro-inflammatory mediators involved in inflammatory mechanisms, which may further alter the normal structure of β-cells by inducing pancreatic islet's apoptosis. In conclusion, it is suggested that the role of inflammation in pathogenesis of T2DM is crucial and cannot be neglected. Moreover, the insight of inflammatory responses in T2DM may provide a new gateway for the better treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Muhammad Sajid Hamid Akash
- Institute of Pharmacology, Toxicology, and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | |
Collapse
|
111
|
Kadam RS, Ramamoorthy P, LaFlamme DJ, McKinsey TA, Kompella UB. Hypoxia alters ocular drug transporter expression and activity in rat and calf models: implications for drug delivery. Mol Pharm 2013; 10:2350-61. [PMID: 23607566 PMCID: PMC3973437 DOI: 10.1021/mp3007133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Chronic hypoxia, a key stimulus for neovascularization, has been implicated in the pathology of proliferative diabetic retinopathy, retinopathy of prematurity, and wet age related macular degeneration. The aim of the present study was to determine the effect of chronic hypoxia on drug transporter mRNA expression and activity in ocular barriers. Sprague-Dawley rats were exposed to hypobaric hypoxia (PB = 380 mmHg) for 6 weeks, and neonatal calves were maintained under hypobaric hypoxia (PB = 445 mmHg) for 2 weeks. Age matched controls for rats, and calves were maintained at ambient altitude and normoxia. The effect of hypoxia on transporter expression was analyzed by qRT-PCR analysis of transporter mRNA expression in hypoxic and control rat choroid-retina. The effect of hypoxia on the activity of PEPT, OCT, ATB(0+), and MCT transporters was evaluated using in vitro transport studies of model transporter substrates across calf cornea and sclera-choroid-RPE (SCRPE). Quantitative gene expression analysis of 84 transporters in rat choroid-retina showed that 29 transporter genes were up regulated or down regulated by ≥1.5-fold in hypoxia. Nine ATP binding cassette (ABC) families of efflux transporters including MRP3, MRP4, MRP5, MRP6, MRP7, Abca17, Abc2, Abc3, and RGD1562128 were up-regulated. For solute carrier family transporters, 11 transporters including SLC10a1, SLC16a3, SLC22a7, SLC22a8, SLC29a1, SLC29a2, SLC2a1, SLC3a2, SLC5a4, SLC7a11, and SLC7a4 were up regulated, while 4 transporters including SLC22a2, SLC22a9, SLC28a1, and SLC7a9 were down-regulated in hypoxia. Of the three aquaporin (Aqp) water channels, Aqp-9 was down-regulated, and Aqp-1 was up-regulated during hypoxia. Gene expression analysis showed down regulation of OCT-1, OCT-2, and ATB(0+) and up regulation of MCT-3 in hypoxic rat choroid-retina, without any effect on the expression of PEPT-1 and PEPT-2. Functional activity assays of PEPT, OCT, ATB(0+), and MCT transporters in calf ocular tissues showed that PEPT, OCT, and ATB(0+) functional activity was down-regulated, whereas MCT functional activity was up-regulated in hypoxic cornea and SCRPE. Gene expression analysis of these transporters in rat tissues was consistent with the functional transport assays except for PEPT transporters. Chronic hypoxia results in significant alterations in the mRNA expression and functional activity of solute transporters in ocular tissues.
Collapse
Affiliation(s)
- Rajendra S. Kadam
- Pharmaceutical Sciences and Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | | | - Timothy A. McKinsey
- Division of Cardiology and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Uday B. Kompella
- Pharmaceutical Sciences and Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
112
|
Maruotti N, Annese T, Cantatore FP, Ribatti D. Macrophages and angiogenesis in rheumatic diseases. Vasc Cell 2013; 5:11. [PMID: 23725043 PMCID: PMC3680215 DOI: 10.1186/2045-824x-5-11] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/26/2013] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis plays a key role in several rheumatic diseases, including rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, systemic sclerosis, systemic lupus erythematosus, and vasculitides. An imbalance between angiogenic inducers and inhibitors seems to be a critical factor in pathogenesis of these diseases. Macrophages promote angiogenesis during rheumatoid arthritis. In addition, macrophages can produce a variety of pro-angiogenic factors that have been associated with the angiogenic response occurring during other rheumatic diseases. Lastly, macrophages could be a target in the treatment of rheumatoid arthritis and other rheumatic diseases. Nevertheless, further studies are needed to better elucidate the exact role of macrophage in angiogenesis in these diseases.
Collapse
Affiliation(s)
- Nicola Maruotti
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School- Ospedale “ D’Avanzo”, Foggia, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Piazza Giulio Cesare, 11, Policlinico, 70124, Bari, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School- Ospedale “ D’Avanzo”, Foggia, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Piazza Giulio Cesare, 11, Policlinico, 70124, Bari, Italy
| |
Collapse
|
113
|
Lim CS, Kiriakidis S, Sandison A, Paleolog EM, Davies AH. Hypoxia-inducible factor pathway and diseases of the vascular wall. J Vasc Surg 2013; 58:219-30. [PMID: 23643279 DOI: 10.1016/j.jvs.2013.02.240] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 02/11/2013] [Accepted: 02/16/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Hypoxia may contribute to the pathogenesis of various diseases of the vascular wall. Hypoxia-inducible factors (HIFs) are nuclear transcriptional factors that regulate the transcription of genes that mediate cellular and tissue homeostatic responses to altered oxygenation. This article reviews the published literature on and discusses the role of the HIF pathway in diseases involving the vascular wall, including atherosclerosis, arterial aneurysms, pulmonary hypertension, vascular graft failure, chronic venous diseases, and vascular malformation. METHODS PubMed was searched with the terms "hypoxia-inducible factor" or "HIF" and "atherosclerosis," "carotid stenosis," "aneurysm," "pulmonary artery hypertension," "varicose veins," "venous thrombosis," "graft thrombosis," and "vascular malformation." RESULTS In atherosclerotic plaque, HIF-1α was localized in macrophages and smooth muscle cells bordering the necrotic core. Increased HIF-1α may contribute to atherosclerosis through alteration of smooth muscle cell proliferation and migration, angiogenesis, and lipid metabolism. The expression of HIF-1α is significantly elevated in aortic aneurysms compared with nonaneurysmal arteries. In pulmonary hypertension, HIF-1α contributes to the increase of intracellular K(+) and Ca(2+) leading to vasoconstriction of pulmonary smooth muscle cells. Alteration of the HIF pathway may contribute to vascular graft failure through the formation of intimal hyperplasia. In chronic venous disease, HIF pathway dysregulation contributes to formation of varicose veins and venous thromboembolism. However, whether the activation of the HIF pathway is protective or destructive to the venous wall is unclear. Increased activation of the HIF pathway causes aberrant expression of angiogenic factors contributing to the formation and maintenance of vascular malformations. CONCLUSIONS Pathologic vascular wall remodelling of many common diseases of the blood vessels has been found to be associated with altered activity of the HIF pathway. Therefore, understanding the role of the HIF pathway in diseases of the vascular wall is important to identify novel therapeutic strategies in the management of these pathologies.
Collapse
Affiliation(s)
- Chung S Lim
- Academic Section of Vascular Surgery, Department of Surgery and Cancer, Faculty of Medicine, Charing Cross Hospital Campus, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
114
|
Cell-free carrier system for localized delivery of peripheral blood cell-derived engineered factor signaling: towards development of a one-step device for autologous angiogenic therapy. J Control Release 2013; 169:91-102. [PMID: 23603614 DOI: 10.1016/j.jconrel.2013.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/07/2013] [Accepted: 04/10/2013] [Indexed: 12/13/2022]
Abstract
Spatiotemporally-controlled delivery of hypoxia-induced angiogenic factor mixtures has been identified by this group as a promising strategy for overcoming the limited ability of chronically ischemic tissues to generate adaptive angiogenesis. We previously developed an implantable, as well as an injectable system for delivering fibroblast-produced factors in vivo. Here, we identify peripheral blood cells (PBCs) as the ideal factor-providing candidates, due to their autologous nature, ease of harvest and ample supply, and investigate wound-simulating biochemical and biophysical environmental parameters that can be controlled to optimize PBC angiogenic activity. It was found that hypoxia (3% O₂) significantly affected the expression of a range of angiogenesis-related factors including VEGF, angiogenin and thrombospondin-1, relative to the normoxic baseline. While all three factors underwent down-regulation over time under hypoxia, there was significant variation in the temporal profile of their expression. VEGF expression was also found to be dependent on cell-scaffold material composition, with fibrin stimulating production the most, followed by collagen and polystyrene. Cell-scaffold matrix stiffness was an additional important factor, as shown by higher VEGF protein levels when PBCs were cultured on stiff vs. compliant collagen hydrogel scaffolds. Engineered PBC-derived factor mixtures could be harvested within cell-free gel and microsphere carriers. The angiogenic effectiveness of factor-loaded carriers could be demonstrated by the ability of their releasates to induce endothelial cell tubule formation and directional migration in in vitro Matrigel assays, and microvessel sprouting in the aortic ring assay. To aid the clinical translation of this approach, we propose a device design that integrates this system, and enables one-step harvesting and delivering of angiogenic factor protein mixtures from autologous peripheral blood. This will facilitate the controlled release of these factors both at the bed-side, as an angiogenic therapy in wounds and peripheral ischemic tissue, as well as pre-, intra- and post-operatively as angiogenic support for central ischemic tissue, grafts, flaps and tissue engineered implants.
Collapse
|
115
|
|
116
|
Muthana M, Rodrigues S, Chen YY, Welford A, Hughes R, Tazzyman S, Essand M, Morrow F, Lewis CE. Macrophage delivery of an oncolytic virus abolishes tumor regrowth and metastasis after chemotherapy or irradiation. Cancer Res 2013; 73:490-5. [PMID: 23172310 DOI: 10.1158/0008-5472.can-12-3056] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Frontline anticancer therapies such as chemotherapy and irradiation often slow tumor growth, but tumor regrowth and spread to distant sites usually occurs after the conclusion of treatment. We recently showed that macrophages could be used to deliver large quantities of a hypoxia-regulated, prostate-specific oncolytic virus (OV) to prostate tumors. In the current study, we show that administration of such OV-armed macrophages 48 hours after chemotherapy (docetaxel) or tumor irradiation abolished the posttreatment regrowth of primary prostate tumors in mice and their spread to the lungs for up to 27 or 40 days, respectively. It also significantly increased the lifespan of tumor-bearing mice compared with those given docetaxel or irradiation alone. These new findings suggest that such a novel, macrophage-based virotherapy could be used to markedly increase the efficacy of chemotherapy and irradiation in patients with prostate cancer.
Collapse
Affiliation(s)
- Munitta Muthana
- Academic Unit of Inflammation & Tumor Targeting, University of Sheffield Medical School, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Manoel-Caetano FS, Xavier DJ, Evangelista AF, Takahashi P, Collares CV, Puthier D, Foss-Freitas MC, Foss MC, Donadi EA, Passos GA, Sakamoto-Hojo ET. Gene expression profiles displayed by peripheral blood mononuclear cells from patients with type 2 diabetes mellitus focusing on biological processes implicated on the pathogenesis of the disease. Gene 2012; 511:151-60. [DOI: 10.1016/j.gene.2012.09.090] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 06/07/2012] [Accepted: 09/11/2012] [Indexed: 12/20/2022]
|
118
|
Mamlouk S, Wielockx B. Hypoxia-inducible factors as key regulators of tumor inflammation. Int J Cancer 2012; 132:2721-9. [PMID: 23055435 DOI: 10.1002/ijc.27901] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/25/2012] [Indexed: 12/23/2022]
Abstract
Low levels of oxygen or hypoxia is often an obstacle in health, particularly in pathological disorders like cancer. The main family of transcription factors responsible for cell survival and adaptation under strenuous conditions of hypoxia are the "hypoxia-inducible factors" (HIFs). Together with prolyl hydroxylase domain enzymes (PHDs), HIFs regulates tumor angiogenesis, proliferation, invasion, metastasis, in addition to resistance to radiation and chemotherapy. Additionally, the entire HIF transcription cascade is involved in the "seventh" hallmark of cancer; inflammation. Studies have shown that hypoxia can influence tumor associated immune cells toward assisting in tumor proliferation, differentiation, vessel growth, distant metastasis and suppression of the immune response via cytokine expression alterations. These changes are not necessarily analogous to HIF's role in non-cancer immune responses, where hypoxia often encourages a strong inflammatory response.
Collapse
Affiliation(s)
- Soulafa Mamlouk
- Emmy Noether Research Group and Institute of Pathology, University of Technology, Dresden, Germany
| | | |
Collapse
|
119
|
Schouppe E, De Baetselier P, Van Ginderachter JA, Sarukhan A. Instruction of myeloid cells by the tumor microenvironment: Open questions on the dynamics and plasticity of different tumor-associated myeloid cell populations. Oncoimmunology 2012; 1:1135-1145. [PMID: 23170260 PMCID: PMC3494626 DOI: 10.4161/onci.21566] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The versatility and plasticity of myeloid cell polarization/differentiation has turned out to be crucial in health and disease, and has become the subject of intense investigation during the last years. On one hand, myeloid cells provide a critical contribution to tissue homeostasis and repair. On the other hand, myeloid cells not only play an important role as first line defense against pathogens but also they are involved in a broad array of inflammation-related diseases such as cancer. Recent studies show that macrophages can exist in different activation states within the same tumor, underlining their plasticity and heterogeneity. In this review, we will discuss recent evidence on how the tumor microenvironment, as it evolves, shapes the recruitment, function, polarization and differentiation of the myeloid cell compartment, leading to the selection of myeloid cells with immunosuppressive and angiogenic functions that facilitate tumor progression and dissemination.
Collapse
Affiliation(s)
- Elio Schouppe
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium
- Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| | - Patrick De Baetselier
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium
- Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| | - Jo A. Van Ginderachter
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium
- Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| | - Adelaida Sarukhan
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium
- Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
- INSERM; Paris, France
| |
Collapse
|
120
|
Leiherer A, Mündlein A, Drexel H. Phytochemicals and their impact on adipose tissue inflammation and diabetes. Vascul Pharmacol 2012; 58:3-20. [PMID: 22982056 DOI: 10.1016/j.vph.2012.09.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 09/03/2012] [Accepted: 09/04/2012] [Indexed: 01/17/2023]
Abstract
Type 2 diabetes mellitus is an inflammatory disease and the mechanisms that underlie this disease, although still incompletely understood, take place in the adipose tissue of obese subjects. Concurrently, the prevalence of obesity caused by Western diet's excessive energy intake and the lack of exercise escalates, and is believed to be causative for the chronic inflammatory state in adipose tissue. Overnutrition itself as an overload of energy may induce the adipocytes to secrete chemokines activating and attracting immune cells to adipose tissue. But also inflammation-mediating food ingredients like saturated fatty acids are believed to directly initiate the inflammatory cascade. In addition, hypoxia in adipose tissue as a direct consequence of obesity, and its effect on gene expression in adipocytes and surrounding cells in fat tissue of obese subjects appears to play a central role in this inflammatory response too. In contrast, revisiting diet all over the world, there are also some natural food products and beverages which are associated with curative effects on human health. Several natural compounds known as spices such as curcumin, capsaicin, and gingerol, or secondary plant metabolites catechin, resveratrol, genistein, and quercetin have been reported to provide an improved health status to their consumers, especially with regard to diabetes, and therefore have been investigated for their anti-inflammatory effect. In this review, we will give an overview about these phytochemicals and their role to interfere with inflammatory cascades in adipose tissue and their potential for fighting against inflammatory diseases like diabetes as investigated in vivo.
Collapse
Affiliation(s)
- Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment, Feldkirch, Austria
| | | | | |
Collapse
|
121
|
Protective effect of Ornithogalum saundersiae Ait (Liliaceae) against acetaminophen-induced acute liver in-jury via CYP2E1 and HIF-1α. Chin J Nat Med 2012. [DOI: 10.3724/sp.j.1009.2012.00177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
122
|
Chiang CS, Fu SY, Wang SC, Yu CF, Chen FH, Lin CM, Hong JH. Irradiation promotes an m2 macrophage phenotype in tumor hypoxia. Front Oncol 2012; 2:89. [PMID: 22888475 PMCID: PMC3412458 DOI: 10.3389/fonc.2012.00089] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/19/2012] [Indexed: 11/16/2022] Open
Abstract
Macrophages display different phenotypes with distinct functions and can rapidly respond to environmental changes. Previous studies on TRAMP-C1 tumor model have shown that irradiation has a strong impact on tumor microenvironments. The major changes include the decrease of microvascular density, the increase of avascular hypoxia, and the aggregation of tumor-associated macrophages in avascular hypoxic regions. Similar changes were observed no matter the irradiation was given to tissue bed before tumor implantation (pre-IR tumors), or to established tumors (IR tumors). Recent results on three murine tumors, TRAMP-C1 prostate adenocarcinoma, ALTS1C1 astrocytoma, and GL261 glioma, further demonstrate that different phenotypes of inflammatory cells are spatially distributed into different microenvironments in both IR and pre-IR tumors. Regions with avascular hypoxia and central necrosis have CD11bhigh/Gr-1+ neutrophils in the center of the necrotic area. Next to them are CD11blow/F4/80+ macrophages that sit at the junctions between central necrotic and surrounding hypoxic regions. The majority of cells in the hypoxic regions are CD11blow/CD68+ macrophages. These inflammatory cell populations express different levels of Arg I. This distribution pattern, except for neutrophils, is not observed in tumors receiving chemotherapy or an anti-angiogenesis agent which also lead to avascular hypoxia. This unique distribution pattern of inflammatory cells in IR tumor sites is interfered with by targeting the expression of a chemokine protein, SDF-1α, by tumor cells, and this also increases radiation-induced tumor growth delay. This indicates that irradiated-hypoxia tissues have distinct tumor microenvironments that favor the development of M2 macrophages and that is affected by the levels of tumor-secreted SDF-1α.
Collapse
Affiliation(s)
- Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University Hsinchu, Taiwan
| | | | | | | | | | | | | |
Collapse
|
123
|
Escribese MM, Casas M, Corbí AL. Influence of low oxygen tensions on macrophage polarization. Immunobiology 2012; 217:1233-40. [PMID: 22889513 DOI: 10.1016/j.imbio.2012.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 07/03/2012] [Accepted: 07/13/2012] [Indexed: 02/05/2023]
Abstract
Microenvironmental conditions in infected, inflamed or damaged tissues are characterized by low levels of oxygen (hypoxia) and nutrients. Myeloid cells (mostly macrophages and neutrophils) account for 95% of the cells newly recruited into inflammatory sites, and exert their effector functions under these restrictive conditions. In the case of macrophages, adaptation to the surrounding tissue environment is underlined by their huge metabolic and functional plasticity, which allows them to critically participate in the maintenance of tissue homeostasis and the initiation and resolution of inflammatory processes under hypoxic conditions. Therefore, alterations in oxygen availability directly affect the macrophage functional state (polarization), a phenomenon that has been already illustrated in pathologies like cancer, atherosclerosis and obesity. This review summarizes recent advances on the molecular basis of macrophage sensing and response to changes in oxygen pressure, emphasizing the link among the hypoxia-induced signalling pathways, macrophage polarization and inflammatory pathologies.
Collapse
Affiliation(s)
- María M Escribese
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain.
| | | | | |
Collapse
|
124
|
Druyan S, Levi E. Reduced O2 concentration during CAM development--its effect on angiogenesis and gene expression in the broiler embryo CAM. Gene Expr Patterns 2012; 12:236-44. [PMID: 22609957 DOI: 10.1016/j.gep.2012.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 04/05/2012] [Accepted: 05/08/2012] [Indexed: 01/07/2023]
Abstract
Hypoxia during embryogenesis may induce changes in the development of some physiological regulatory systems, thereby causing permanent phenotypic changes in the embryo. Various levels of hypoxia at different time points during embryogenesis were found to affect both anatomical and physiological morphogenesis. These changes and adaptations depended on the timing, intensity, and duration of the hypoxic exposure and, moreover, were regulated by differential expression of developmentally important genes, mostly expressed in a stage- and time-dependent manner. Eggs incubated in a 17%-oxygen atmosphere for 12 h/d from E5 through E12 exhibited a clear and significant increase in the vascular area of the chorioallantoic membrane (CAM); an increase that was already significant within 12 h after the end of the 1st hypoxic exposures (E6). We used the combination of the genes, β-actin, RPLP0 and HPRT as a reference for gene expression profiling, in studying the expression levels of hypoxia-inducible factor 1-alpha (HIF1α), vascular endothelial growth factor alpha-2 (VEGF α 2), vascular endothelial growth factor receptor 2 (KDR), matrix metalloproteinase-2 (MMP2), and fibroblast growth factor 2 (FGF2), under normal and hypoxic conditions. In general, expression of all five investigated genes throughout the embryonic day of development had similar patterns of hypoxia-induced alterations. In E5.5 embryos, expression of HIF1α, MMP2, VEGFα2, and KDR was significantly higher in hypoxic embryos than in controls. In E6 embryos expression of HIF1α, VEGFα2, and FGF2 was significantly higher in hypoxic embryos than in controls. From E6.5 onward expression levels of the examined genes did not show any differences between hypoxic and control embryos. It can be concluded that in this experimental model, exposing broiler embryos to 17% O(2) from E5 to E7 induced significant angiogenesis, as expressed by the above genes. Further studies to examine whether this early exposure to hypoxic condition affects the chick's ability to withstand a post-hatch hypoxic environment is still required.
Collapse
Affiliation(s)
- S Druyan
- Institute of Animal Science, ARO, the Volcani Center, P.O. Box 6, Bet Dagan 50250, Israel.
| | | |
Collapse
|
125
|
Shen Z, Seppänen H, Vainionpää S, Ye Y, Wang S, Mustonen H, Puolakkainen P. IL10, IL11, IL18 are differently expressed in CD14+ TAMs and play different role in regulating the invasion of gastric cancer cells under hypoxia. Cytokine 2012; 59:352-7. [PMID: 22595646 DOI: 10.1016/j.cyto.2012.04.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 04/22/2012] [Indexed: 01/13/2023]
Abstract
BACKGROUND Recent evidence shows that chronic inflammation mediated by tumor-associated macrophage (TAM) play an important role in malignant tumor formation and progression. Interleukins expressed in TAMs regulate this progress. Hypoxia is a salient feature of solid tumors and has a profound influence on the biology of TAMs However, the role of interleukins in the gastric cancer progression under hypoxia is not clear. METHODS Realtime RT-PCR was used to quantitatively investigate the IL10, IL11 and IL18 expression in CD14(-) normal macrophages and CD14(+) TAMs co-cultured with four gastric cancer cell lines including non-metastatic cell line AGS and metastatic cell lines HGC-27, Hs-746T and NCI-N87 under normal or hypoxic conditions. In addition, the correlation between IL10, IL11, IL18 expression in TAMs under hypoxia and mobility of gastric cancer cells were analyzed. RESULTS Under normal conditions, the IL10 and IL18 expressions were significantly higher in CD14(+) TAMs co-cultured with non-metastatic cell line than with metastatic cell lines. IL11 expression was significantly higher in CD14(+) TAMs co-cultured with distant metastasis cell lines. Hypoxia induced IL10, IL11 and IL18 expression up regulated significantly in TAMs co-cultured with AGS, Hs-746T and NCI-N87 cell line. There was a significant negative correlation between IL11 expression in CD14(+) TAMs and gastric cancer cell invasion speed under hypoxic conditions (r=0.861, P<0.001). CONCLUSION The up-regulation of IL10, IL11 and IL18 expression in TAMs by hypoxia differed in gastric cancer cell lines. IL11 expression in TAMs might play more important role than IL10 and IL18 expression in regulating the invasion of gastric cancer cells under hypoxia.
Collapse
Affiliation(s)
- Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, PR China.
| | | | | | | | | | | | | |
Collapse
|
126
|
Meda C, Molla F, De Pizzol M, Regano D, Maione F, Capano S, Locati M, Mantovani A, Latini R, Bussolino F, Giraudo E. Semaphorin 4A exerts a proangiogenic effect by enhancing vascular endothelial growth factor-A expression in macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 188:4081-92. [PMID: 22442441 DOI: 10.4049/jimmunol.1101435] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The axon guidance cues semaphorins (Semas) and their receptors plexins have been shown to regulate both physiological and pathological angiogenesis. Sema4A plays an important role in the immune system by inducing T cell activation, but to date, the role of Sema4A in regulating the function of macrophages during the angiogenic and inflammatory processes remains unclear. In this study, we show that macrophage activation by TLR ligands LPS and polyinosinic-polycytidylic acid induced a time-dependent increase of Sema4A and its receptors PlexinB2 and PlexinD1. Moreover, in a thioglycollate-induced peritonitis mouse model, Sema4A was detected in circulating Ly6C(high) inflammatory monocytes and peritoneal macrophages. Acting via PlexinD1, exogenous Sema4A strongly increased macrophage migration. Of note, Sema4A-activated PlexinD1 enhanced the expression of vascular endothelial growth factor-A, but not of inflammatory chemokines. Sema4A-stimulated macrophages were able to activate vascular endothelial growth factor receptor-2 and the PI3K/serine/threonine kinase Akt pathway in endothelial cells and to sustain their migration and in vivo angiogenesis. Remarkably, in an in vivo cardiac ischemia/reperfusion mouse model, Sema4A was highly expressed in macrophages recruited at the injured area. We conclude that Sema4A activates a specialized and restricted genetic program in macrophages able to sustain angiogenesis and participates in their recruitment and activation in inflammatory injuries.
Collapse
Affiliation(s)
- Claudia Meda
- Department of Oncological Sciences, University of Torino School of Medicine, 10060 Candiolo, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Mogulkoc U, Coskunpinar E, Aynaci E, Cağlar E, Ortakoylu MG, Ozkan G, Oltulu YM, Eraltan IY. Is MMP-7 gene polymorphism a possible risk factor for chronic obstructive pulmonary disease in Turkish patients. Genet Test Mol Biomarkers 2012; 16:519-23. [PMID: 22417142 DOI: 10.1089/gtmb.2011.0271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease that leads to fixed narrowing of small airways and alveolar wall destruction (emphysema). This study was performed to test the association between MMP-7 (rs155668818) and MMP-12 (rs56184183) polymorphisms in the MMP-7 gene and COPD risk and its severity in the Turkish population. MMP-7 and MMP-12 polymorphisms were genotyped in 85 patients with COPD and 73 healthy control subjects using real-time polymerase chain reaction analysis. There were significant differences in the distribution of MMP-7 genotypes but not in the frequencies of these alleles between COPD patients and controls (p=0.009, p=0.102, respectively). The MMP-7 AA genotype was found to be associated with an increased risk of COPD (p=0.004; odds ratio: 2.576; confidence interval: 1.297-5.119). The lowest values of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC), FEV1/FVC in patients with GG homozygosity were determined and these values were statistically significant compared to the control subjects (p<0.001, p<0.001, p<0.001). When the present study groups were analyzed for MMP-12 polymorphism, it was found that all the subjects had wild-type genotype for this polymorphism. These findings have suggested that MMP-7 polymorphism might be associated with the risk and progression of COPD in the Turkish population.
Collapse
Affiliation(s)
- Umit Mogulkoc
- Yedikule Chest Diseases and Chest Surgery, Training and Research Hospital, Istanbul, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Kadam SD, Gucek M, Cole RN, Watkins PA, Comi AM. Cell proliferation and oxidative stress pathways are modified in fibroblasts from Sturge-Weber syndrome patients. Arch Dermatol Res 2012; 304:229-35. [PMID: 22402795 DOI: 10.1007/s00403-012-1210-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 01/03/2012] [Accepted: 01/09/2012] [Indexed: 01/05/2023]
Abstract
Sturge-Weber syndrome (SWS) is defined by vascular malformations of the face, eye and brain and an underlying somatic mutation has been hypothesized. We employed isobaric tags for relative and absolute quantification (iTRAQ-8plex)-based liquid chromatography interfaced with tandem mass spectrometry (LC-MS/MS) approach to identify differentially expressed proteins between port-wine-derived and normal skin-derived fibroblasts of four individuals with SWS. Proteins were identified that were significantly up- or down-regulated (i.e., ratios >1.2 or <0.8) in two or three pairs of samples (n = 31/972 quantified proteins) and their associated p values reported. Ingenuity pathway analysis (IPA) tool showed that the up-regulated proteins were associated with pathways that enhance cell proliferation; down-regulated proteins were associated with suppression of cell proliferation. The significant toxicologic list pathway in all four observations was oxidative stress mediated by Nrf2. This proteomics study highlights oxidative stress also consistent with a possible mutation in the RASA1 gene or pathway in SWS.
Collapse
Affiliation(s)
- Shilpa D Kadam
- Departments of Neurology and Developmental Medicine, Hugo Moser Kennedy Krieger Research Institute, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
129
|
Chouaib S, Messai Y, Couve S, Escudier B, Hasmim M, Noman MZ. Hypoxia promotes tumor growth in linking angiogenesis to immune escape. Front Immunol 2012; 3:21. [PMID: 22566905 PMCID: PMC3341970 DOI: 10.3389/fimmu.2012.00021] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 02/04/2012] [Indexed: 01/01/2023] Open
Abstract
Despite the impressive progress over the past decade, in the field of tumor immunology, such as the identification of tumor antigens and antigenic peptides, there are still many obstacles in eliciting an effective immune response to eradicate cancer. It has become increasingly clear that tumor microenvironment plays a crucial role in the control of immune protection. Tumors have evolved to utilize hypoxic stress to their own advantage by activating key biochemical and cellular pathways that are important in progression, survival, and metastasis. Hypoxia-inducible factor (HIF-1) and vascular endothelial growth factor (VEGF) play a determinant role in promoting tumor cell growth and survival. Hypoxia contributes to immune suppression by activating HIF-1 and VEGF pathways. Accumulating evidence suggests a link between hypoxia and tumor tolerance to immune surveillance through the recruitment of regulatory cells (regulatory T cells and myeloid derived suppressor cells). In this regard, hypoxia (HIF-1α and VEGF) is emerging as an attractive target for cancer therapy. How the microenvironmental hypoxia poses both obstacles and opportunities for new therapeutic immune interventions will be discussed.
Collapse
Affiliation(s)
- Salem Chouaib
- INSERM U753, Institut Gustave Roussy Villejuif, France
| | | | | | | | | | | |
Collapse
|
130
|
Baze MM, Hunter K, Hayes JP. Chronic hypoxia stimulates an enhanced response to immune challenge without evidence of an energetic tradeoff. ACTA ACUST UNITED AC 2012; 214:3255-68. [PMID: 21900473 DOI: 10.1242/jeb.054544] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is broad interest in whether there is a tradeoff between energy metabolism and immune function, and how stress affects immune function. Under hypoxic stress, maximal aerobic metabolism is limited, and other aspects of energy metabolism of animals may be altered as well. Although acute hypoxia appears to enhance certain immune responses, the effects of chronic hypoxia on immune function are largely unstudied. We tested: (1) whether chronic hypoxia affects immune function and (2) whether hypoxia affects the metabolic cost of immune function. First, flow cytometry was used to monitor the peripheral blood immunophenotype of mice over the course of 36 days of hypoxic exposure. Second, hypoxic and normoxic mice were subjected to an adaptive immune challenge via keyhole limpet hemocyanin (KLH) or to an innate immune challenge via lipopolysaccharide (LPS). The resting metabolic rates of mice in all immune challenge treatments were also measured. Although hypoxia had little effect on the peripheral blood immunophenotype, hypoxic mice challenged with KLH or LPS had enhanced immunological responses in the form of higher antibody titers or increased TNF-α production, respectively. Initially, mice exposed to hypoxia had lower metabolic rates, but this response was transitory and resting metabolic rates were normal by the end of the experiment. There was no effect of either immune challenge on resting metabolic rate, suggesting that mounting either the acute phase response or a humoral response is not as energetically expensive as previously thought. In addition, our results suggest that immune responses to chronic and acute hypoxia are concordant. Both forms of hypoxia appear to stimulate both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Monica M Baze
- Program in Ecology, Evolution and Conservation Biology and Department of Biology, University of Nevada, Reno, NV 89557, USA.
| | | | | |
Collapse
|
131
|
|
132
|
Motz GT, Coukos G. The parallel lives of angiogenesis and immunosuppression: cancer and other tales. Nat Rev Immunol 2011; 11:702-11. [DOI: 10.1038/nri3064] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
133
|
An HJ, Shin H, Jo SG, Kim YJ, Lee JO, Paik SG, Lee H. The survival effect of mitochondrial Higd-1a is associated with suppression of cytochrome C release and prevention of caspase activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:2088-98. [PMID: 21856340 DOI: 10.1016/j.bbamcr.2011.07.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 06/27/2011] [Accepted: 07/27/2011] [Indexed: 12/20/2022]
Abstract
Higd-1a (hypoxia induced gene domain family-1a) is a mitochondrial inner membrane protein with a conformation of N-terminal outside-C-terminal outside and loop inside. There are four Higd genes, Higd-1a, -1b, -1c and -2a, in the mouse. Higd-1a and -2a are expressed primarily in the brain, heart, kidney and leukocytes. HIF (hypoxia-inducible factor) overexpression induced the endogenous expression and promoter activity of Higd-1a. Mutation of the HRE (hypoxia-response element) site at -32bp in the Higd-1a promoter reduced the promoter activity, suggesting that transcription of Higd-1a is regulated by binding of the transcription factor HIF to the HRE. Higd-1a promoted cell survival under hypoxia. RAW264.7 cells stably transfected with Higd-1a underwent less apoptosis than control cells in a hypoxic condition, and hypoxia-induced apoptosis was strongly enhanced when endogenous Higd-1a was silenced by siRNA. The survival effect of Higd-1a was completely abolished by deletion of the 26 N-terminal amino acids, and we showed that Higd-1a increased survival by inhibiting cytochrome C release and reducing the activities of caspases. However, expression of Bcl-2, Bax, Bad, and BNIP3 and translocation of AIF were unaffected under the same conditions. Higd-2a also enhanced cell survival under hypoxia. Cells transfected with Higd-2a underwent less apoptosis than control cells in hypoxic conditions, and hypoxia-induced apoptosis increased when endogenous Higd-2a was depleted. Together these observations indicate that Higd-1a is induced by hypoxia in a HIF-dependent manner and its anti-apoptotic effect results from inhibiting cytochrome C release and reducing caspase activities.
Collapse
Affiliation(s)
- Hyun-Jung An
- Department of Biology, BK21 Daedeok R&D Innopolis Bio Brain Center, Chungnam National University, Daejeon, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
134
|
Expression of Genes Encoding Extracellular Matrix Macromolecules and Metalloproteinases in Avian Tibial Dyschondroplasia. J Comp Pathol 2011; 145:174-86. [DOI: 10.1016/j.jcpa.2010.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/23/2010] [Accepted: 12/13/2010] [Indexed: 01/27/2023]
|
135
|
|
136
|
Seo GM, Rachakatla RS, Balivada S, Pyle M, Shrestha TB, Basel MT, Myers C, Wang H, Tamura M, Bossmann SH, Troyer DL. A self-contained enzyme activating prodrug cytotherapy for preclinical melanoma. Mol Biol Rep 2011; 39:157-65. [PMID: 21567204 DOI: 10.1007/s11033-011-0720-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 04/23/2011] [Indexed: 02/01/2023]
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) has been investigated as a means of cancer treatment without affecting normal tissues. This system is based on the delivery of a suicide gene, a gene encoding an enzyme which is able to convert its substrate from non-toxic prodrug to cytotoxin. In this experiment, we have developed a targeted suicide gene therapeutic system that is completely contained within tumor-tropic cells and have tested this system for melanoma therapy in a preclinical model. First, we established double stable RAW264.7 monocyte/macrophage-like cells (Mo/Ma) containing a Tet-On® Advanced system for intracellular carboxylesterase (InCE) expression. Second, we loaded a prodrug into the delivery cells, double stable Mo/Ma. Third, we activated the enzyme system to convert the prodrug, irinotecan, to the cytotoxin, SN-38. Our double stable Mo/Ma homed to the lung melanomas after 1 day and successfully delivered the prodrug-activating enzyme/prodrug package to the tumors. We observed that our system significantly reduced tumor weights and numbers as targeted tumor therapy after activation of the InCE. Therefore, we propose that this system may be a useful targeted melanoma therapy system for pulmonary metastatic tumors with minimal side effects, particularly if it is combined with other treatments.
Collapse
Affiliation(s)
- Gwi-Moon Seo
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Bench TJ, Jeremias A, Brown DL. Matrix metalloproteinase inhibition with tetracyclines for the treatment of coronary artery disease. Pharmacol Res 2011; 64:561-6. [PMID: 21624471 DOI: 10.1016/j.phrs.2011.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Coronary artery disease is caused by atherosclerosis - a progressive arterial inflammatory disease that is responsible for significant global mortality and morbidity through the development of the acute coronary syndromes: sudden cardiac death, acute myocardial infarction and unstable angina. These clinical entities share a common pathophysiology: rupture of atherosclerotic plaque resulting in abrupt complete or partial thrombotic obstruction of coronary blood flow. Matrix metalloproteinases (MMP), through their central role in tissue remodeling and inflammation, are secreted by inflammatory cells of the atherosclerotic plaque and are capable of degrading all the extracellular matrix components of the fibrous cap that separates the atherosclerotic lesion from blood flow in the arterial lumen. Plaque rupture occurs when the circumferential tensile stresses in the artery overwhelm the structural integrity of the progressively degraded, thinned and weakened fibrous cap of the atherosclerotic lesion. Tetracyclines inhibit MMPs through their ability to chelate zinc. Subantimicrobial doses of doxycycline have been shown to reduce inflammation and inhibit MMP activity in patients with coronary artery disease. Further investigation is warranted to assess the potential clinical risks and benefits of MMP inhibition with tetracyclines or other agents in the treatment of coronary artery disease.
Collapse
Affiliation(s)
- Travis J Bench
- Department of Medicine (Cardiovascular Medicine), Stony Brook University, Health Sciences Center T 16-080, Stony Brook, NY 11794-8171, USA
| | | | | |
Collapse
|
138
|
Tomita S, Kihira Y, Imanishi M, Fukuhara Y, Imamura Y, Ishizawa K, Ikeda Y, Tsuchiya K, Tamaki T. Pathophysiological response to hypoxia - from the molecular mechanisms of malady to drug discovery:inflammatory responses of hypoxia-inducible factor 1α (HIF-1α) in T cells observed in development of vascular remodeling. J Pharmacol Sci 2011; 115:433-9. [PMID: 21422726 DOI: 10.1254/jphs.10r22fm] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Recent studies have shown that the cellular immune response to the hypoxic microenvironment constructed by vascular remodeling development modulates the resulting pathologic alterations. A major mechanism mediating adaptive responses to reduced oxygen availability is the regulation of transcription by hypoxia-inducible factor 1 (HIF-1). Impairment of HIF-1-dependent inflammatory responses in T cells causes an augmented vascular remodeling induced by arterial injury, which is shown as prominent neointimal hyperplasia and increase in infiltration of inflammatory cells at the adventitia in mice lacking Hif-1α specifically in T cells. Studies to clarify the mechanism of augmented vascular remodeling in the mutant mice have shown enhanced production of cytokines in activated T cells and augmented antibody production in response to a T-dependent antigen in the mutant mice. This minireview shows that HIF-1α in T cells plays a crucial role in vascular inflammation and remodeling in response to cuff injury as a negative regulator of the T cell-mediated immune response and suggests potential new therapeutic strategies that target HIF-1α.
Collapse
Affiliation(s)
- Shuhei Tomita
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Ong LL, Li W, Oldigs JK, Kaminski A, Gerstmayer B, Piechaczek C, Wagner W, Li RK, Ma N, Steinhoff G. Hypoxic/normoxic preconditioning increases endothelial differentiation potential of human bone marrow CD133+ cells. Tissue Eng Part C Methods 2011; 16:1069-81. [PMID: 20073989 DOI: 10.1089/ten.tec.2009.0641] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD133+ cells are hemangioblasts that have capacity to generate into both hematopoietic and endothelial cells (ECs). Hypoxia/normoxia has shown to be the regulator of the balance between stemness and differentiation. In this study we performed Agilent's whole human genome oligo microarray analysis and examined the differentiation potential of the bone-marrow-derived CD133+ cells after hypoxic/normoxic preconditioning of CD133+ cells. Results showed that there was no significant increase in erythroid colony forming unit (CFU-E) and CFU-granulocyte, erythrocyte, monocyte, and megakaryocyte formation with cells treated under hypoxia/normoxia. However, a significant increment of EC forming unit at 24 h (143.2 +/- 8.0%) compared to 0 h (100 +/- 11.4%) was observed in CFU-EC analysis. Reverse transcription-polymerase chain reaction and immunostaining analysis showed that the differentiated cells diminished hematopoietic stem cell surface markers and acquired the gene markers and functional phenotype of ECs. The transcriptome profile revealed a cluster of 232 downregulated and 498 upregulated genes in cells treated for 24 h under hypoxia. The upregulated genes include angiogenic genes, angiogenic growth factor genes, angiogenic cytokine and chemokine genes, as well as angiogenic-positive regulatory genes, including FGFBP1, PDGFB, CCL15, CXCL12, CXCL6, IL-6, PTN, EREG, ERBB2, EDG5, FGF3, FHF2, GDF15, JUN, L1CAM, NRG1, NGFR, and PDGFB. On the other hand, angiogenesis inhibitors and related genes, including IL12A, MLLT7, STAB1, and TIMP2, are downregulated. Taken together, hypoxic/normoxic preconditioning may lead to the differentiation of CD133+ cells toward endothelial lineage, which may improve the current clinical trial studies.
Collapse
Affiliation(s)
- Lee-Lee Ong
- Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Abstract
Components of the immune system are altered in obesity and type 2 diabetes (T2D), with the most apparent changes occurring in adipose tissue, the liver, pancreatic islets, the vasculature and circulating leukocytes. These immunological changes include altered levels of specific cytokines and chemokines, changes in the number and activation state of various leukocyte populations and increased apoptosis and tissue fibrosis. Together, these changes suggest that inflammation participates in the pathogenesis of T2D. Preliminary results from clinical trials with salicylates and interleukin-1 antagonists support this notion and have opened the door for immunomodulatory strategies for the treatment of T2D that simultaneously lower blood glucose levels and potentially reduce the severity and prevalence of the associated complications of this disease.
Collapse
|
141
|
Muthana M, Giannoudis A, Scott SD, Fang HY, Coffelt SB, Morrow FJ, Murdoch C, Burton J, Cross N, Burke B, Mistry R, Hamdy F, Brown NJ, Georgopoulos L, Hoskin P, Essand M, Lewis CE, Maitland NJ. Use of Macrophages to Target Therapeutic Adenovirus to Human Prostate Tumors. Cancer Res 2011; 71:1805-15. [DOI: 10.1158/0008-5472.can-10-2349] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
142
|
Lynch CC. Matrix metalloproteinases as master regulators of the vicious cycle of bone metastasis. Bone 2011; 48:44-53. [PMID: 20601294 DOI: 10.1016/j.bone.2010.06.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 06/07/2010] [Accepted: 06/09/2010] [Indexed: 01/29/2023]
Abstract
Bone remodeling is a delicate balancing act between the bone matrix synthesizing osteoblasts and bone resorbing osteoclasts. Active bone metastases typically subvert this process to generate lesions that are comprised of extensive areas of pathological osteogenesis and osteolysis. The resultant increase in bone matrix remodeling enhances cytokine/growth factor bioavailability thus creating a vicious cycle that stimulates tumor progression. Given the extent of matrix remodeling occurring in the tumor-bone microenvironment, the expression of matrix metalloproteinases (MMPs) would be expected, since collectively they have the ability to degrade all components of the extracellular matrix (ECM). However, in addition to being "matrix bulldozers", MMPs control the bioavailability and bioactivity of factors such as RANKL and TGFβ that have been described as crucial for tumor-bone interaction, thus implicating MMPs as key regulators of the vicious cycle of bone metastases.
Collapse
Affiliation(s)
- Conor C Lynch
- Department of Orthopaedics and Rehabilitation, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
143
|
Biomechanics and Pathobiology of Aortic Aneurysms. STUDIES IN MECHANOBIOLOGY, TISSUE ENGINEERING AND BIOMATERIALS 2011. [DOI: 10.1007/8415_2011_84] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
144
|
Staples KJ, Sotoodehnejadnematalahi F, Pearson H, Frankenberger M, Francescut L, Ziegler-Heitbrock L, Burke B. Monocyte-derived macrophages matured under prolonged hypoxia transcriptionally up-regulate HIF-1α mRNA. Immunobiology 2010; 216:832-9. [PMID: 21281980 DOI: 10.1016/j.imbio.2010.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
This study tested the hypothesis that prolonged severe hypoxia during monocyte to macrophage differentiation results in macrophages with a pattern of gene expression and phenotype distinct from those maturing in normal oxygen levels. Macrophages accumulate in hypoxic and anoxic areas within pathological sites such as tumours, wounds, and arthritic joints, and have been proposed as vehicles for gene therapy delivery to such tissues. Several non-pathological tissues are also hypoxic. We therefore argue that differentiation from monocyte to macrophage in hypoxic conditions is a common occurrence. However, the effect of long term severe hypoxia on monocyte to macrophage differentiation has not been studied. Here, using primary human peripheral blood monocytes, we show that maturation for 5 days in 0.2% oxygen results in decreased phagocytosis, and decreased CD40 and CD206 expression. Chronic hypoxia induced much higher mRNA levels of the pro-angiogenic cytokine, VEGF, in adherence-purified macrophages (27-fold), CD14-magnetic bead purified monocytes (90-fold), and PBMC (104-fold) compared to acute (24h) hypoxia (11, 17 and 9-fold, respectively). This suggests that macrophages may play an even greater role in angiogenesis than previously appreciated. Furthermore, chronic hypoxia resulted in up-regulation of HIF-1α mRNA, in all monocyte-derived macrophage types studied. Actinomycin D experiments indicate that the increases in HIF-1α mRNA were not due to increased mRNA stability. To our knowledge this is the first study demonstrating up-regulation of HIF-1α mRNA by hypoxia in macrophages. Taken together, the data support the hypothesis that hypoxia affects monocyte to macrophage maturation, resulting in a distinct gene expression pattern and phenotype.
Collapse
Affiliation(s)
- Karl J Staples
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, UK
| | | | | | | | | | | | | |
Collapse
|
145
|
Garcia-Albeniz X, Pericay C, Alonso-Espinaco V, Alonso V, Escudero P, Fernández-Martos C, Gallego R, Gascón P, Castellví-Bel S, Maurel J. Serum matrilysin correlates with poor survival independently of KRAS and BRAF status in refractory advanced colorectal cancer patients treated with irinotecan plus cetuximab. Tumour Biol 2010; 32:417-24. [PMID: 21104178 DOI: 10.1007/s13277-010-0136-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 11/15/2010] [Indexed: 01/05/2023] Open
Abstract
The purpose of the study was to prospectively explore the role of serum MMP-7 as a predictive and prognostic marker of anti-epidermal growth factor receptor (EGFR) therapy and irinotecan efficacy in third-line advanced colorectal cancer therapy. One hundred patients were recruited prospectively from six Spanish hospitals. Patients were treated with biweekly irinotecan 180 mg/m(2) and cetuximab 400 mg/m(2) (loading dose) and weekly cetuximab 250 mg/m(2) until progressive disease or unacceptable toxicity. Baseline MMP-7 was determined using a quantitative solid-phase sandwich ELISA. KRAS and BRAF mutational status were also assessed. The clinical endpoints examined were overall survival (OS), progression-free survival (PFS), and response rate. No association between serum MMP-7 and neither KRAS nor BRAF mutational status was found. The multivariate analysis revealed that MMP-7 predicts PFS both in wild-type (WT) KRAS patients (HR 1.03, 95% CI 1.00-1.06; p = 0.046) and in mutant KRAS patients (HR 1.18, 95% CI 1.01-1.35; p = 0.036). The presence of mutant BRAF was associated with shorter PFS (HR 8.49, 95% CI 2.88-25.0; p < 0.001) and worse OS (HR 3.55, 95% CI 1.39-9.09; p = 0.008) in the subset of WT KRAS patients. Serum MMP-7 is associated with PFS in colorectal patients treated with anti-EGFR therapy as third-line treatment independently of KRAS status.
Collapse
Affiliation(s)
- Xabier Garcia-Albeniz
- Medical Oncology Department, Institut Clínic Malalties Hemato-Oncológiques (ICMHO), Hospital Clínic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERehd, Villarroel 170, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Allen M, Louise Jones J. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J Pathol 2010; 223:162-76. [PMID: 21125673 DOI: 10.1002/path.2803] [Citation(s) in RCA: 256] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/01/2010] [Accepted: 10/01/2010] [Indexed: 12/13/2022]
Abstract
It is now recognized that the host microenvironment undergoes extensive change during the evolution and progression of cancer. This involves the generation of cancer-associated fibroblasts (CAFs), which, through release of growth factors and cytokines, lead to enhanced angiogenesis, increased tumour growth and invasion. It has also been demonstrated that CAFs may modulate the cancer stem cell (CSC) phenotype, which has therapeutic implications. The altered fibroblast phenotype also contributes to the development of an altered extracellular matrix (ECM), with synthesis of ECM isoforms rarely found in normal tissues, including tenascin-C isoforms and the fibronectin EDA isoform. There is also emerging evidence of how the tensile strength of the tumour-associated ECM may be modified and lead to altered signalling in tumour cells. The hypoxic environment of the tumour stimulates angiogenesis and also impacts on other aspects of cell signalling, including the c-met pathway and lysyl oxidase-mediated signalling, which can directly promote tumour cell invasion. The inflammatory infiltrate associated with many solid tumours also modulates tumour function, having both anti- and pro-tumour effects. All of these components of the microenvironment provide potential targets for therapeutic attack, with a number of molecules already in clinical trials. It is also becoming evident that characterizing the tumour microenvironment can provide important prognostic and predictive information about tumours, independent of the tumour cell phenotype.
Collapse
Affiliation(s)
- Michael Allen
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, London, UK
| | | |
Collapse
|
147
|
Grant MM, Kolamunne RT, Lock FE, Matthews JB, Chapple ILC, Griffiths HR. Oxygen tension modulates the cytokine response of oral epithelium to periodontal bacteria. J Clin Periodontol 2010; 37:1039-48. [PMID: 20955352 DOI: 10.1111/j.1600-051x.2010.01622.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND There is an inverse relationship between pocket depth and pocket oxygen tension with deep pockets being associated with anaerobic bacteria. However, little is known about how the host tissues respond to bacteria under differing oxygen tensions within the periodontal pocket. AIM To investigate the effect of different oxygen tensions upon nuclear factor-kappa B (NF-κB) activation and the inflammatory cytokine response of oral epithelial cells when exposed to nine species of oral bacteria. MATERIALS AND METHODS H400 oral epithelial cells were equilibrated at 2%, 10% or 21% oxygen. Cells were stimulated with heat-killed oral bacteria at multiplicity of infection 10:1, Escherichia coli lipopolysaccharide (15 μg/ml) or vehicle control. Interleukin-8 (IL-8) and tumour necrosis factor-alpha (TNF-α) levels were measured by enzyme-linked immunosorbent assay and NF-κB activation was measured by reporter vector or by immunohistochemical analysis. RESULTS Tannerella forsythensis, Porphyromonas gingivalis and Prevotella intermedia elicited the greatest epithelial NF-κB activation and cytokine responses. An oxygen-tension-dependent trend in cytokine production was observed with the highest IL-8 and TNF-α production observed at 2% oxygen and lowest at 21% oxygen. CONCLUSIONS These data demonstrate a greater pro-inflammatory host response and cell signalling response to bacteria present in more anaerobic conditions, and hypersensitivity of epithelial cells to pro-inflammatory stimuli at 2% oxygen, which may have implications for disease pathogenesis and/or therapy.
Collapse
Affiliation(s)
- Melissa M Grant
- Periodontal Research Group, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | | | | | | | | | | |
Collapse
|
148
|
Losser MR, Damoisel C, Payen D. Bench-to-bedside review: Glucose and stress conditions in the intensive care unit. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:231. [PMID: 20727232 PMCID: PMC2945096 DOI: 10.1186/cc9100] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The physiological response to blood glucose elevation is the pancreatic release of insulin, which blocks hepatic glucose production and release, and stimulates glucose uptake and storage in insulin-dependent tissues. When this first regulatory level is overwhelmed (that is, by exogenous glucose supplementation), persistent hyperglycaemia occurs with intricate consequences related to the glucose acting as a metabolic substrate and as an intracellular mediator. It is thus very important to unravel the glucose metabolic pathways that come into play during stress as well as the consequences of these on cellular functions. During acute injuries, activation of serial hormonal and humoral responses inducing hyperglycaemia is called the 'stress response'. Central activation of the nervous system and of the neuroendocrine axes is involved, releasing hormones that in most cases act to worsen the hyperglycaemia. These hormones in turn induce profound modifications of the inflammatory response, such as cytokine and mediator profiles. The hallmarks of stress-induced hyperglycaemia include 'insulin resistance' associated with an increase in hepatic glucose output and insufficient release of insulin with regard to glycaemia. Although both acute and chronic hyperglycaemia may induce deleterious effects on cells and organs, the initial acute endogenous hyperglycaemia appears to be adaptive. This acute hyperglycaemia participates in the maintenance of an adequate inflammatory response and consequently should not be treated aggressively. Hyperglycaemia induced by an exogenous glucose supply may, in turn, amplify the inflammatory response such that it becomes a disproportionate response. Since chronic exposure to glucose metabolites, as encountered in diabetes, induces adverse effects, the proper roles of these metabolites during acute conditions need further elucidation.
Collapse
Affiliation(s)
- Marie-Reine Losser
- Laboratoire de Recherche Paris 7 EA 3509, Service d'Anesthésie-Réanimation, Hôpital Lariboisière, Assistance Publique - Hôpitaux de Paris, Université Diderot Paris-7, 75475 Paris Cedex 10, France.
| | | | | |
Collapse
|
149
|
Cleavage of E-Cadherin by Matrix Metalloproteinase-7 Promotes Cellular Proliferation in Nontransformed Cell Lines via Activation of RhoA. JOURNAL OF ONCOLOGY 2010; 2010:530745. [PMID: 20628524 PMCID: PMC2902104 DOI: 10.1155/2010/530745] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 04/01/2010] [Indexed: 01/08/2023]
Abstract
Perturbations in cell-cell contact machinery occur frequently in epithelial cancers and result in increased cancer cell migration and invasion. Previously, we demonstrated that MMP-7, a protease implicated in mammary and intestinal tumor growth, can process the adherens junction component E-cadherin. This observation leads us to test whether MMP-7 processing of E-cadherin could directly impact cell proliferation in nontransformed epithelial cell lines (MDCK and C57MG). Our goal was to investigate the possibility that MMP-7 produced by cancer cells may have effects on adjacent normal epithelium. Here, we show that MMP-7 processing of E-cadherin mediates, (1) loss of cell-cell contact, (2) increased cell migration, (3) a loss of epithelial cell polarization and (4) increased cell proliferation via RhoA activation. These data demonstrate that MMP-7 promotes epithelial cell proliferation via the processing of E-cadherin and provide insights into the molecular mechanisms that govern epithelial cell growth.
Collapse
|
150
|
Jiang H, Zhu YS, Xu H, Sun Y, Li QF. Inflammatory stimulation and hypoxia cooperatively activate HIF-1α in bronchial epithelial cells: involvement of PI3K and NF-κB. Am J Physiol Lung Cell Mol Physiol 2010; 298:L660-9. [PMID: 20139176 DOI: 10.1152/ajplung.00394.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The transcription factor hypoxia-inducible factor (HIF)-1 plays a central physiological role in oxygen and energy homeostasis, and is activated during hypoxia by stabilization of the subunit HIF-1α. Recent studies have demonstrated that non-hypoxic stimuli can also activate HIF-1α in a cell-specific manner. Here, we demonstrate that stimulation of BEAS-2B cells and primary human bronchial epithelial cells by proinflammatory cytokines TNFα/IL-4 strongly induced expression and transcriptional activity of HIF-1α under normoxic conditions and amplified hypoxic HIF-1α activation. TNFα/IL-4 stimulated de novo HIF-1α gene transcription and translation rather than affected HIF-1α protein degradation and mRNA decay process. The activation of HIF-1α by TNFα/IL-4 was countered by the phosphoinositol 3-kinase (PI3K) inhibitor LY-294002 and rapamycin, an antagonist of mammalian target of rapamycin (mTOR), but not by inhibition of the MAPK pathway. In line, TNFα/IL-4 also activated NF-κB, whereas blocking of NF-κB by an inhibitor or silencing NF-κB subunit p65 attenuated HIF-1α activation by TNFα/IL-4. We also found the collaborative induction of VEGF, a potent angiogenic factor required for airway remodeling, by TNFα/IL-4 and hypoxia partially via HIF-1α pathway in BEAS-2B cells. This study reports the previously unsuspected collaborative regulation of HIF-1α by TNFα/IL-4 and hypoxia in bronchial epithelial cells partially via PI3K-mTOR and NF-κB pathway, and thereby will lead to the elucidation of the importance of HIF-1 in integrating inflammatory and hypoxic response in the pathogenesis of airway diseases.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Sen Zhu
- Department of Anesthesiology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Xu
- Department of Anesthesiology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Fang Li
- Department of Anesthesiology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|