51
|
Ow CPC, Ngo JP, Ullah MM, Hilliard LM, Evans RG. Renal hypoxia in kidney disease: Cause or consequence? Acta Physiol (Oxf) 2018; 222:e12999. [PMID: 29159875 DOI: 10.1111/apha.12999] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/10/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Tissue hypoxia has been proposed as an important factor in the pathophysiology of both chronic kidney disease (CKD) and acute kidney injury (AKI), initiating and propagating a vicious cycle of tubular injury, vascular rarefaction, and fibrosis and thus exacerbation of hypoxia. Here, we critically evaluate this proposition by systematically reviewing the literature relevant to the following six questions: (i) Is kidney disease always associated with tissue hypoxia? (ii) Does tissue hypoxia drive signalling cascades that lead to tissue damage and dysfunction? (iii) Does tissue hypoxia per se lead to kidney disease? (iv) Does tissue hypoxia precede pathology? (v) Does tissue hypoxia colocalize with pathology? (vi) Does prevention of tissue hypoxia prevent kidney disease? We conclude that tissue hypoxia is a common feature of both AKI and CKD. Furthermore, at least under in vitro conditions, renal tissue hypoxia drives signalling cascades that lead to tissue damage and dysfunction. Tissue hypoxia itself can lead to renal pathology, independent of other known risk factors for kidney disease. There is also some evidence that tissue hypoxia precedes renal pathology, at least in some forms of kidney disease. However, we have made relatively little progress in determining the spatial relationships between tissue hypoxia and pathological processes (i.e. colocalization) or whether therapies targeted to reduce tissue hypoxia can prevent or delay the progression of renal disease. Thus, the hypothesis that tissue hypoxia is a "common pathway" to both AKI and CKD still remains to be adequately tested.
Collapse
Affiliation(s)
- C. P. C. Ow
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - J. P. Ngo
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - M. M. Ullah
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - L. M. Hilliard
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - R. G. Evans
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| |
Collapse
|
52
|
Yang WJ, Hao YX, Yang X, Fu XL, Shi Y, Yue HL, Yin P, Dong HL, Yu PW. Overexpression of Tie2 is associated with poor prognosis in patients with gastric cancer. Oncol Lett 2018; 15:8027-8033. [PMID: 29849805 DOI: 10.3892/ol.2018.8329] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 03/14/2017] [Indexed: 12/21/2022] Open
Abstract
Tunica Interna endothelial cell kinase (Tie2)-expressing macrophages (TEMs) are a subgroup of tumor-associated macrophages that are associated with a poor prognosis in numerous types of cancer. The present study aimed to assess the prognostic impact of Tie2 expression in gastric cancer tissues. Between January 2009 and December 2009, 76 newly diagnosed patients with gastric cancer at the Southwest Hospital, Third Military Medical University (Chongqing, China) were enrolled. TEMs were detected using immunohistochemistry. Tie2, cluster of differentiation (CD)68 and carbonic anhydrase IX (CAIX) were analyzed using immunohistochemistry and immunofluorescent microscopy. Tie2 protein expression was analyzed using western blot analysis in hypoxic and normoxic gastric cancer tissues. The number of TEMs positively staining for Tie2 increased with the tumor-node-metastasis (TNM) stage: 0, 53.9, 75.6 and 100% in stages I, II, III and IV, respectively (P<0.001). Tumor size and lymph node involvement were significantly associated with the presence of Tie2 in the tumor stroma (P<0.001). There was no significant difference between Tie2 and CAIX, irrespective of how the patients were grouped (tumor size, lymph node involvement, TNM stage or histological grade). Tie2 protein expression was increased in the hypoxic regions of gastric tumors.Tie2 and CD68 expression colocalized in hypoxic and normoxic gastric cancer tissues. The 1-, 2- and 3-year recurrence rates of the TEM-positive group were 31.4, 56.9 and 66.7%, respectively, as compared with 8, 28 and 48%, respectively, for the TEM-negative group (P<0.05). In the TEM-negative group, 2 patients succumbed to the disease, as compared with 21 patients in the TEM-positive group (P<0.05). Therefore, high quantities of TEMs, represented by Tie2 expression, in gastric tumors may be associated with poor survival.
Collapse
Affiliation(s)
- Wei-Jun Yang
- Department of General Surgery and Center of Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China.,Department of General Surgery, The First People's Hospital of Guiyang, Guiyang, Guizhou 550002, P.R. China
| | - Ying-Xue Hao
- Department of General Surgery and Center of Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xia Yang
- Department of Immunology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiao-Lan Fu
- Department of Immunology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yan Shi
- Department of General Surgery and Center of Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Hai-Ling Yue
- Department of General Surgery and Center of Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Peng Yin
- Department of General Surgery and Center of Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Hao-Lin Dong
- Department of Immunology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Pei-Wu Yu
- Department of General Surgery and Center of Minimally Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
53
|
Mohamed E, Al-Khami AA, Rodriguez PC. The cellular metabolic landscape in the tumor milieu regulates the activity of myeloid infiltrates. Cell Mol Immunol 2018; 15:421-427. [PMID: 29568118 DOI: 10.1038/s41423-018-0001-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 12/26/2022] Open
Abstract
Malignant cells upregulate distinct energy metabolism programs that support their proliferation, migration, and adaptation to the stressful tumor microenvironment (TME). Additionally, this exaggerated metabolic activity allows cancer cells to hijack essential nutrients and outcompete neighboring infiltrating immune cells, thereby impairing antitumor immunity. During recent years, there has been great interest in the field to understand the tumor-induced energy metabolism signals that regulate the function of immune cells in individuals with cancer. Accordingly, it is now well accepted that uncovering the mechanisms that instruct the metabolic behavior of cancer cells and tumor-associated immune cells is an indispensable strategy for the development of new approaches to overcome immune suppression in tumors. Thus, in this minireview, we briefly discuss the interaction between particular metabolic signaling pathways and immunosuppressive activity in different subsets of myeloid cells within the TME. Additionally, we illustrate potential central mechanisms controlling the metabolic reprogramming of myeloid cells in response to tumor-derived factors.
Collapse
Affiliation(s)
- Eslam Mohamed
- H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr. MRC-Annex 2nd, Tampa, FL, 33612, USA
| | - Amir A Al-Khami
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Paulo C Rodriguez
- H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr. MRC-Annex 2nd, Tampa, FL, 33612, USA.
| |
Collapse
|
54
|
Hypoxia promotes glioma-associated macrophage infiltration via periostin and subsequent M2 polarization by upregulating TGF-beta and M-CSFR. Oncotarget 2018; 7:80521-80542. [PMID: 27602954 PMCID: PMC5348338 DOI: 10.18632/oncotarget.11825] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 08/13/2016] [Indexed: 01/08/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are enriched in gliomas and help create a tumor-immunosuppressive microenvironment. A distinct M2-skewed type of macrophages makes up the majority of glioma TAMs, and these cells exhibit pro-tumor functions. Gliomas contain large hypoxic areas, and the presence of a correlation between the density of M2-polarized TAMs and hypoxic areas suggests that hypoxia plays a supportive role during TAM recruitment and induction. Here, we investigated the effects of hypoxia on human macrophage recruitment and M2 polarization. We also investigated the influence of the HIF inhibitor acriflavine (ACF) on M2 TAM infiltration and tumor progression in vivo. We found that hypoxia increased periostin (POSTN) expression in glioma cells and promoted the recruitment of macrophages. Hypoxia-inducible POSTN expression was increased by TGF-α via the RTK/PI3K pathway, and this effect was blocked by treating hypoxic cells with ACF. We also demonstrated that both a hypoxic environment and hypoxia-treated glioma cell supernatants were capable of polarizing macrophages toward a M2 phenotype. ACF partially reversed the M2 polarization of macrophages by inhibiting the upregulation of M-CSFR in macrophages and TGF-β in glioma cells under hypoxic conditions. Administering ACF also ablated tumor progression in vivo. Our findings reveal a mechanism that underlies hypoxia-induced TAM enrichment and M2 polarization and suggest that pharmacologically inhibiting HIFs may reduce M2-polarized TAM infiltration and glioma progression.
Collapse
|
55
|
Aryaeian N, Sedehi SK, Arablou T. Polyphenols and their effects on diabetes management: A review. Med J Islam Repub Iran 2017; 31:134. [PMID: 29951434 PMCID: PMC6014790 DOI: 10.14196/mjiri.31.134] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Indexed: 12/15/2022] Open
Abstract
Background: Type 2 diabetes is a growing public health problem and is associated with increased morbidity and mortality. The worldwide prevalence of type 2 diabetes is rising. Polyphenols, such as flavonoids, phenolic acid, and stilbens, are a large and heterogeneous group of phytochemicals in plant-based foods. In this review, we aimed at assessing the studies on polyphenols and diabetes management. Methods: A literature search in the PubMed, EMBASE, Scopus, and ISI Web of Science databases was conducted to identify relevant studies published from 1986 to Jan 2017. Results: Several animal models and a limited number of human studies have revealed that polyphenols decrease hyperglycemia and improve acute insulin secretion and insulin sensitivity. The possible mechanisms include decrease in glucose absorption in the intestine, inhibition of carbohydrates digestion, stimulation of insulin secretion, modulation of glucose release from the liver, activation of insulin receptors and glucose uptake in insulin-sensitive tissues, modulation of intracellular signaling pathways, and gene expression. Conclusion: Growing evidence indicates that various dietary polyphenols may influence blood glucose at different levels and may also help control and prevent diabetes complication. However, we still need more clinical trials to determine the effects of polyphenols- rich foods, their effective dose, and mechanisms of their effects in managing diabetes.
Collapse
Affiliation(s)
- Naheed Aryaeian
- Research Center for Environmental Health Technology, Iran University of Medical Sciences and Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Khorshidi Sedehi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Arablou
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
56
|
Kong F, Liu Z, Jain VG, Shima K, Suzuki T, Muglia LJ, Starczynowski DT, Pasare C, Bhattacharyya S. Inhibition of IRAK1 Ubiquitination Determines Glucocorticoid Sensitivity for TLR9-Induced Inflammation in Macrophages. THE JOURNAL OF IMMUNOLOGY 2017; 199:3654-3667. [PMID: 29038250 DOI: 10.4049/jimmunol.1700443] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/19/2017] [Indexed: 01/26/2023]
Abstract
Inflammatory responses are controlled by signaling mediators that are regulated by various posttranslational modifications. Recently, transcription-independent functions for glucocorticoids (GC) in restraining inflammation have emerged, but the underlying mechanisms are unknown. In this study, we report that GC receptor (GR)-mediated actions of GC acutely suppress TLR9-induced inflammation via inhibition of IL-1R-associated kinase 1 (IRAK1) ubiquitination. β-TrCP-IRAK1 interaction is required for K48-linked ubiquitination of IRAK1 at Lys134 and subsequent membrane-to-cytoplasm trafficking of IRAK1 interacting partners TNFR-associated factor 6 and TAK1 that facilitates NF-κB and MAPK activation. Upon costimulation of macrophages with GC and TLR9-engaging ligand, GR physically interacts with IRAK1 and interferes with protein-protein interactions between β-TrCP and IRAK1. Ablation of GR in macrophages prevents GC-dependent suppression of β-TrCP-IRAK1 interactions. This GC-mediated suppression of IRAK1 activation is unique to TLR9, as GC treatment impairs TLR9 but not TLR4 ligand-induced K48-linked IRAK1 ubiquitination and trafficking of IRAK1 interacting partners. Furthermore, mutations in IRAK1 at Lys134 prevent TLR9 ligand-induced activation of inflammatory signaling mediators and synthesis of proinflammatory cytokines to an extent comparable to GC-mediated inhibition. Collectively, these findings identify a transcription-independent, rapid, and nongenomic GC suppression of TLR9 ligand-mediated IRAK1 ubiquitination as a novel mechanism for restraining acute inflammatory reactions.
Collapse
Affiliation(s)
- Fansheng Kong
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Zhiwei Liu
- Neonatal Division, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Viral G Jain
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Kenjiro Shima
- Division of Pulmonary Biology, Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Takuji Suzuki
- Division of Pulmonary Biology, Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Louis J Muglia
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Daniel T Starczynowski
- Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Chandrashekhar Pasare
- Department of Immunology, Southwestern Medical Center, University of Texas, Dallas, TX 75390
| | - Sandip Bhattacharyya
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229;
| |
Collapse
|
57
|
Ng CF, Frieboes HB. Model of vascular desmoplastic multispecies tumor growth. J Theor Biol 2017; 430:245-282. [PMID: 28529153 PMCID: PMC5614902 DOI: 10.1016/j.jtbi.2017.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 03/07/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022]
Abstract
We present a three-dimensional nonlinear tumor growth model composed of heterogeneous cell types in a multicomponent-multispecies system, including viable, dead, healthy host, and extra-cellular matrix (ECM) tissue species. The model includes the capability for abnormal ECM dynamics noted in tumor development, as exemplified by pancreatic ductal adenocarcinoma, including dense desmoplasia typically characterized by a significant increase of interstitial connective tissue. An elastic energy is implemented to provide elasticity to the connective tissue. Cancer-associated fibroblasts (myofibroblasts) are modeled as key contributors to this ECM remodeling. The tumor growth is driven by growth factors released by these stromal cells as well as by oxygen and glucose provided by blood vasculature which along with lymphatics are stimulated to proliferate in and around the tumor based on pro-angiogenic factors released by hypoxic tissue regions. Cellular metabolic processes are simulated, including respiration and glycolysis with lactate fermentation. The bicarbonate buffering system is included for cellular pH regulation. This model system may be of use to simulate the complex interactions between tumor and stromal cells as well as the associated ECM and vascular remodeling that typically characterize malignant cancers notorious for poor therapeutic response.
Collapse
Affiliation(s)
- Chin F Ng
- Department of Bioengineering, University of Louisville, Lutz Hall 419, KY 40208, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Lutz Hall 419, KY 40208, USA; James Graham Brown Cancer Center, University of Louisville, KY, USA.
| |
Collapse
|
58
|
Rabold K, Netea MG, Adema GJ, Netea-Maier RT. Cellular metabolism of tumor-associated macrophages - functional impact and consequences. FEBS Lett 2017; 591:3022-3041. [PMID: 28771701 DOI: 10.1002/1873-3468.12771] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 12/20/2022]
Abstract
Macrophages are innate immune cells that play a role not only in host defense against infections, but also in the pathophysiology of autoimmune and autoinflammatory disorders, as well as cancer. An important feature of macrophages is their high plasticity, with high ability to adapt to environmental changes by adjusting their cellular metabolism and immunological phenotype. Macrophages are one of the most abundant innate immune cells within the tumor microenvironment that have been associated with tumor growth, metastasis, angiogenesis and poor prognosis. In the context of cancer, however, so far little is known about metabolic changes in macrophages, which have been shown to determine functional fate of the cells in other diseases. Here, we review the current knowledge regarding the cellular metabolism of tumor-associated macrophages (TAMs) and discuss its implications for cell function. Understanding the regulation of the cellular metabolism of TAMs may reveal novel therapeutic targets for treatment of malignancies.
Collapse
Affiliation(s)
- Katrin Rabold
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany
| | - Gosse J Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
59
|
Singh Y, Pawar VK, Meher JG, Raval K, Kumar A, Shrivastava R, Bhadauria S, Chourasia MK. Targeting tumor associated macrophages (TAMs) via nanocarriers. J Control Release 2017; 254:92-106. [DOI: 10.1016/j.jconrel.2017.03.395] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
60
|
Martinez-Marin D, Jarvis C, Nelius T, de Riese W, Volpert OV, Filleur S. PEDF increases the tumoricidal activity of macrophages towards prostate cancer cells in vitro. PLoS One 2017; 12:e0174968. [PMID: 28403150 PMCID: PMC5389654 DOI: 10.1371/journal.pone.0174968] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/19/2017] [Indexed: 12/23/2022] Open
Abstract
Background Although inflammation and prostate cancer (PCa) have been linked, the molecular interactions between macrophages and PCa cells are poorly explored. Pigment Epithelium-Derived Factor (PEDF) is an anti-angiogenic and anti-tumor factor. We previously showed that PEDF induces macrophages recruitment in vitro, correlates with macrophages density in human prostate, and stimulates macrophages polarization towards the classically activated pathway. Here, we demonstrate that PEDF modulates the interaction between macrophages and PCa cells through a bidirectional signalling leading to tumor cell apoptosis and phagocytosis. Methods RAW 264.7 and THP-1 cells, and BMDMs were grown in vitro as mono- or co-cultures with PC3 or CL1 tumor cells. The effects of PEDF and its derived P18 peptide were measured on macrophages differentiation, migration, and superoxide production, and tumor cell apoptosis and phagocytosis. PEDF receptors (ATP5B, PNPLA2, and LRP6) and CD47 mRNA and protein expression were quantified in macrophages and tumor cells by quantitative RT-PCR, western blot, immunofluorescence and flow cytometry. Results We found that PEDF induced the migration of macrophages towards tumor 3D spheroids and 2D cultures. In co-culture, PEDF increased PCa cells phagocytosis through an indirect apoptosis-dependent mechanism. Moreover, PEDF stimulated the production of superoxide by macrophages. Conditioned media from macrophages exposed to PEDF induced tumor cells apoptosis in contrast to control conditioned media suggesting that ROS may be involved in tumor cells apoptosis. ATP5B and PNPLA2 PEDF receptors on macrophages and CD47 on tumor cells were respectively up- and down-regulated by PEDF. As PEDF, blocking CD47 induced phagocytosis. Inhibiting ATP5B reduced phagocytosis. Inversely, PNPLA2 inhibition blocks differentiation but maintains phagocytosis. CD47-induced phagocytosis was partially reverted by ATP5B inhibition suggesting a complementary action. Similar effects were observed with P18 PEDF-derived peptide. Conclusions These data established that modulating the molecular interactions between macrophages and PCa cells using PEDF may be a promising strategy for PCa treatment.
Collapse
Affiliation(s)
- Dalia Martinez-Marin
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Courtney Jarvis
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Thomas Nelius
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Werner de Riese
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Olga V. Volpert
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Stéphanie Filleur
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
61
|
Lapeyre-Prost A, Terme M, Pernot S, Pointet AL, Voron T, Tartour E, Taieb J. Immunomodulatory Activity of VEGF in Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:295-342. [PMID: 28215534 DOI: 10.1016/bs.ircmb.2016.09.007] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability of tumor cells to escape tumor immunosurveillance contributes to cancer development. Factors produced in the tumor microenvironment create "tolerizing" conditions and thereby help the tumor to evade antitumoral immune responses. VEGF-A, already known for its major role in tumor vessel growth (neoangiogenesis), was recently identified as a key factor in tumor-induced immunosuppression. In particular, VEGF-A fosters the proliferation of immunosuppressive cells, limits T-cell recruitment into tumors, and promotes T-cell exhaustion. Antiangiogenic therapies have shown significant efficacy in patients with a variety of solid tumors, preventing tumor progression by limiting tumor-induced angiogenesis. VEGF-targeting therapies have also been shown to modulate the tumor-induced immunosuppressive microenvironment, enhancing Th1-type T-cell responses and increasing tumor infiltration by T cells. The immunomodulatory properties of VEGF-targeting therapies open up new perspectives for cancer treatment, especially through strategies combining antiangiogenic drugs with immunotherapy. Preclinical models and early clinical studies of these combined approaches have given promising results.
Collapse
Affiliation(s)
- A Lapeyre-Prost
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France
| | - M Terme
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France.
| | - S Pernot
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'hépatogastroentérologie et d'oncologie digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - A-L Pointet
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'hépatogastroentérologie et d'oncologie digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - T Voron
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service de chirurgie digestive, Hôpital Européen Georges Pompidou, Paris, France
| | - E Tartour
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'immunologie biologique. Hôpital Européen Georges Pompidou, Paris, France
| | - J Taieb
- INSERM U970, PARCC (Paris Cardiovascular Research Center), Université Paris-Descartes, Paris, France; Service d'hépatogastroentérologie et d'oncologie digestive, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
62
|
Yang PW, Hsu IJ, Chang CW, Wang YC, Hsieh CY, Shih KH, Wong LF, Shih NY, Hsieh MS, Hou MTK, Lee JM. Visible-absorption spectroscopy as a biomarker to predict treatment response and prognosis of surgically resected esophageal cancer. Sci Rep 2016; 6:33414. [PMID: 27624872 PMCID: PMC5022060 DOI: 10.1038/srep33414] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/30/2016] [Indexed: 12/19/2022] Open
Abstract
The application of optical absorption spectra in prognostic prediction has hardly been investigated. We developed and evaluated a novel two dimensional absorption spectrum measurement system (TDAS) for use in early diagnosis, evaluating response to chemoradiation, and making prognostic prediction. The absorption spectra of 120 sets of normal and tumor tissues from esophageal cancer patients were analyzed with TDAS ex-vivo. We demonstrated the cancerous tissue, the tissue from patients with a poor concurrent chemoradiotherapy (CCRT) response, and the tissue from patients with an early disease progression each had a readily identifiable common spectral signature. Principal component analysis (PCA) classified tissue spectra into distinct groups, demonstrating the feasibility of using absorption spectra in differentiating normal and tumor tissues, and in predicting CCRT response, poor survival and tumor recurrence (efficiencies of 75%, 100% and 85.7% respectively). Multivariate analysis revealed that patients identified as having poor-response, poor-survival and recurrence spectral signatures were correlated with increased risk of poor response to CCRT (P = 0.012), increased risk of death (P = 0.111) and increased risk of recurrence (P = 0.030) respectively. Our findings suggest that optical absorption microscopy has great potential to be a useful tool for pre-operative diagnosis and prognostic prediction of esophageal cancer.
Collapse
Affiliation(s)
- Pei-Wen Yang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - I-Jen Hsu
- Department of Physics and Center for Biomedical Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Chun-Wei Chang
- Department of Physics and Center for Biomedical Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Yu-Chia Wang
- Department of Physics and Center for Biomedical Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Ching-Yueh Hsieh
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Hui Shih
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Li-Fan Wong
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Nai-Yu Shih
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Min-Shu Hsieh
- Graduate Institute of Pathology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Max Ti-Kuang Hou
- Department of Mechanical Engineering, National United University, Miaoli, Taiwan
| | - Jang-Ming Lee
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
63
|
Ciasca G, Papi M, Minelli E, Palmieri V, De Spirito M. Changes in cellular mechanical properties during onset or progression of colorectal cancer. World J Gastroenterol 2016; 22:7203-7214. [PMID: 27621568 PMCID: PMC4997642 DOI: 10.3748/wjg.v22.i32.7203] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/11/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) development represents a multistep process starting with specific mutations that affect proto-oncogenes and tumour suppressor genes. These mutations confer a selective growth advantage to colonic epithelial cells that form first dysplastic crypts, and then malignant tumours and metastases. All these steps are accompanied by deep mechanical changes at the cellular and the tissue level. A growing consensus is emerging that such modifications are not merely a by-product of the malignant progression, but they could play a relevant role in the cancer onset and accelerate its progression. In this review, we focus on recent studies investigating the role of the biomechanical signals in the initiation and the development of CRC. We show that mechanical cues might contribute to early phases of the tumour initiation by controlling the Wnt pathway, one of most important regulators of cell proliferation in various systems. We highlight how physical stimuli may be involved in the differentiation of non-invasive cells into metastatic variants and how metastatic cells modify their mechanical properties, both stiffness and adhesion, to survive the mechanical stress associated with intravasation, circulation and extravasation. A deep comprehension of these mechanical modifications may help scientist to define novel molecular targets for the cure of CRC.
Collapse
|
64
|
Abstract
The anoxemia theory proposes that an imbalance between the demand for and supply of oxygen in the arterial wall is a key factor in the development of atherosclerosis. There is now substantial evidence that there are regions within the atherosclerotic plaque in which profound hypoxia exists; this may fundamentally change the function, metabolism, and responses of many of the cell types found within the developing plaque and whether the plaque will evolve into a stable or unstable phenotype. Hypoxia is characterized in molecular terms by the stabilization of hypoxia-inducible factor (HIF) 1α, a subunit of the heterodimeric nuclear transcriptional factor HIF-1 and a master regulator of oxygen homeostasis. The expression of HIF-1 is localized to perivascular tissues, inflammatory macrophages, and smooth muscle cells adjacent to the necrotic core of atherosclerotic lesions and regulates several genes that are important to vascular function including vascular endothelial growth factor, nitric oxide synthase, endothelin-1, and erythropoietin. This review summarizes the effects of hypoxia on the functions of cells involved in atherogenesis and the evidence for its potential importance from experimental models and clinical studies.
Collapse
Affiliation(s)
- Gordon A A Ferns
- 1 Department of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| | - Lamia Heikal
- 1 Department of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| |
Collapse
|
65
|
Microbial and Natural Metabolites That Inhibit Splicing: A Powerful Alternative for Cancer Treatment. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3681094. [PMID: 27610372 PMCID: PMC5004037 DOI: 10.1155/2016/3681094] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/27/2016] [Accepted: 07/03/2016] [Indexed: 02/06/2023]
Abstract
In eukaryotes, genes are frequently interrupted with noncoding sequences named introns. Alternative splicing is a nuclear mechanism by which these introns are removed and flanking coding regions named exons are joined together to generate a message that will be translated in the cytoplasm. This mechanism is catalyzed by a complex machinery known as the spliceosome, which is conformed by more than 300 proteins and ribonucleoproteins that activate and regulate the precision of gene expression when assembled. It has been proposed that several genetic diseases are related to defects in the splicing process, including cancer. For this reason, natural products that show the ability to regulate splicing have attracted enormous attention due to its potential use for cancer treatment. Some microbial metabolites have shown the ability to inhibit gene splicing and the molecular mechanism responsible for this inhibition is being studied for future applications. Here, we summarize the main types of natural products that have been characterized as splicing inhibitors, the recent advances regarding molecular and cellular effects related to these molecules, and the applications reported so far in cancer therapeutics.
Collapse
|
66
|
Abstract
The role of tumor-associated macrophages (TAMs) in cancer is often correlated with poor prognosis, even though this statement should be interpreted with care, as the effects of macrophages primarily depend on their localization within the tumor. This versatile cell type orchestrates a broad spectrum of biological functions and exerts very complex and even opposing functions on cell death, immune stimulation or suppression, and angiogenesis, resulting in an overall pro- or antitumoral effect. We are only beginning to understand the environmental cues that contribute to transient retention of macrophages in a specific phenotype. It has become clear that hypoxia shapes and induces specific macrophage phenotypes that serve tumor malignancy, as hypoxia promotes immune evasion, angiogenesis, tumor cell survival, and metastatic dissemination. Additionally, TAMs in the hypoxic niches within the tumor are known to mediate resistance to several anticancer treatments and to promote cancer relapse. Thus, a careful characterization and understanding of this macrophage differentiation state is needed in order to efficiently tailor cancer therapy.
Collapse
|
67
|
Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene 2016; 36:439-445. [PMID: 27345407 DOI: 10.1038/onc.2016.225] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 12/11/2022]
Abstract
Tumors use several strategies to evade the host immune response, including creation of an immune-suppressive and hostile tumor environment. Tissue hypoxia due to inadequate blood supply is reported to develop very early during tumor establishment. Hypoxic stress has a strong impact on tumor cell biology. In particular, tissue hypoxia contributes to therapeutic resistance, heterogeneity and progression. It also interferes with immune plasticity, promotes the differentiation and expansion of immune-suppressive stromal cells, and remodels the metabolic landscape to support immune privilege. Therefore, tissue hypoxia has been regarded as a central factor for tumor aggressiveness and metastasis. In this regard, manipulating host-tumor interactions in the context of the hypoxic tumor microenvironment may be important in preventing or reverting malignant conversion. We will discuss how tumor microenvironment-driven transient compositional tumor heterogeneity involves hypoxic stress. Tumor hypoxia is a therapeutic concern since it can reduce the effectiveness of conventional therapies as well as cancer immunotherapy. Thus, understanding how tumor and stromal cells respond to hypoxia will allow for the design of innovative cancer therapies that can overcome these barriers. A better understanding of hypoxia-dependent mechanisms involved in the regulation of immune tolerance could lead to new strategies to enhance antitumor immunity. Therefore, discovery and validation of therapeutic targets derived from the hypoxic tumor microenvironment is of major importance. In this context, critical hypoxia-associated pathways are attractive targets for immunotherapy of cancer. In this review, we summarize current knowledge regarding the molecular mechanisms induced by tumor cell hypoxia with a special emphasis on therapeutic resistance and immune suppression. We emphasize mechanisms of manipulating hypoxic stress and its associated pathways, which may support the development of more durable and successful cancer immunotherapy approaches in the future.
Collapse
|
68
|
Ruddy JM, Ikonomidis JS, Jones JA. Multidimensional Contribution of Matrix Metalloproteinases to Atherosclerotic Plaque Vulnerability: Multiple Mechanisms of Inhibition to Promote Stability. J Vasc Res 2016; 53:1-16. [PMID: 27327039 PMCID: PMC7196926 DOI: 10.1159/000446703] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/07/2016] [Indexed: 12/17/2022] Open
Abstract
The prevalence of atherosclerotic disease continues to increase, and despite significant reductions in major cardiovascular events with current medical interventions, an additional therapeutic window exists. Atherosclerotic plaque growth is a complex integration of cholesterol penetration, inflammatory cell infiltration, vascular smooth muscle cell (VSMC) migration, and neovascular invasion. A family of matrix-degrading proteases, the matrix metalloproteinases (MMPs), contributes to all phases of vascular remodeling. The contribution of specific MMPs to endothelial cell integrity and VSMC migration in atherosclerotic lesion initiation and progression has been confirmed by the increased expression of these proteases in plasma and plaque specimens. Endogenous blockade of MMPs by the tissue inhibitors of metalloproteinases (TIMPs) may attenuate proteolysis in some regions, but the progression of matrix degeneration suggests that MMPs predominate in atherosclerotic plaque, precipitating vulnerability. Plaque neovascularization also contributes to instability and, coupling the known role of MMPs in angiogenesis to that of atherosclerotic plaque growth, interest in targeting MMPs to facilitate plaque stabilization continues to accumulate. This article aims to review the contributions of MMPs and TIMPs to atherosclerotic plaque expansion, neovascularization, and rupture vulnerability with an interest in promoting targeted therapies to improve plaque stabilization and decrease the risk of major cardiovascular events.
Collapse
Affiliation(s)
- Jean Marie Ruddy
- Division of Vascular Surgery, Department of Surgery, Medical University of South Carolina, Charleston, S.C., USA
| | | | | |
Collapse
|
69
|
Boyd AJ, Kuhn DC, Lozowy RJ, Kulbisky GP. Low wall shear stress predominates at sites of abdominal aortic aneurysm rupture. J Vasc Surg 2016; 63:1613-9. [DOI: 10.1016/j.jvs.2015.01.040] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/28/2015] [Indexed: 10/23/2022]
|
70
|
Ordonez AA, Tasneen R, Pokkali S, Xu Z, Converse PJ, Klunk MH, Mollura DJ, Nuermberger EL, Jain SK. Mouse model of pulmonary cavitary tuberculosis and expression of matrix metalloproteinase-9. Dis Model Mech 2016; 9:779-88. [PMID: 27482816 PMCID: PMC4958312 DOI: 10.1242/dmm.025643] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/20/2016] [Indexed: 01/12/2023] Open
Abstract
Cavitation is a key pathological feature of human tuberculosis (TB), and is a well-recognized risk factor for transmission of infection, relapse after treatment and the emergence of drug resistance. Despite intense interest in the mechanisms underlying cavitation and its negative impact on treatment outcomes, there has been limited study of this phenomenon, owing in large part to the limitations of existing animal models. Although cavitation does not occur in conventional mouse strains after infection with Mycobacterium tuberculosis, cavitary lung lesions have occasionally been observed in C3HeB/FeJ mice. However, to date, there has been no demonstration that cavitation can be produced consistently enough to support C3HeB/FeJ mice as a new and useful model of cavitary TB. We utilized serial computed tomography (CT) imaging to detect pulmonary cavitation in C3HeB/FeJ mice after aerosol infection with M. tuberculosis Post-mortem analyses were performed to characterize lung lesions and to localize matrix metalloproteinases (MMPs) previously implicated in cavitary TB in situ A total of 47-61% of infected mice developed cavities during primary disease or relapse after non-curative treatments. Key pathological features of human TB, including simultaneous presence of multiple pathologies, were noted in lung tissues. Optical imaging demonstrated increased MMP activity in TB lesions and MMP-9 was significantly expressed in cavitary lesions. Tissue MMP-9 activity could be abrogated by specific inhibitors. In situ, three-dimensional analyses of cavitary lesions demonstrated that 22.06% of CD11b+ signal colocalized with MMP-9. C3HeB/FeJ mice represent a reliable, economical and tractable model of cavitary TB, with key similarities to human TB. This model should provide an excellent tool to better understand the pathogenesis of cavitation and its effects on TB treatments.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rokeya Tasneen
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Supriya Pokkali
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ziyue Xu
- Center for Infectious Disease Imaging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul J Converse
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mariah H Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel J Mollura
- Center for Infectious Disease Imaging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric L Nuermberger
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
71
|
Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol 2016; 12:385-97. [PMID: 27225300 DOI: 10.1038/nrrheum.2016.69] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
72
|
Giacomelli R, Ruscitti P, Alvaro S, Ciccia F, Liakouli V, Di Benedetto P, Guggino G, Berardicurti O, Carubbi F, Triolo G, Cipriani P. IL-1β at the crossroad between rheumatoid arthritis and type 2 diabetes: may we kill two birds with one stone? Expert Rev Clin Immunol 2016; 12:849-55. [PMID: 26999417 DOI: 10.1586/1744666x.2016.1168293] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although in the past the prevention of joint destruction in rheumatoid arthritis (RA) was strongly emphasized, now a great interest is focused on associated comorbidities in these patients. Multiple data suggest that a large percentage of RA patients are affected by Type 2 Diabetes (T2D), whose incidence has reached epidemic levels in recent years, thus increasing the health care costs. A better knowledge about the pathogenesis of these diseases as well as the mechanisms of action of drugs may allow both policy designers and physicians to choose the most effective treatments, thus lowering the costs. This review will focus on the role of Interleukin (IL)-1β in the pathogenesis of both the diseases, the efficacy of IL-1 blocking molecules in controlling these diseases, and will provide information suggesting that targeting IL-1β, in patients affected by both RA and T2D, may be a promising therapeutic choice.
Collapse
Affiliation(s)
- Roberto Giacomelli
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Piero Ruscitti
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Saverio Alvaro
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Francesco Ciccia
- b Division of Rheumatology, Department of Internal Medicine , University of Palermo , Palermo , Italy
| | - Vasiliki Liakouli
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Paola Di Benedetto
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Giuliana Guggino
- b Division of Rheumatology, Department of Internal Medicine , University of Palermo , Palermo , Italy
| | - Onorina Berardicurti
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Francesco Carubbi
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| | - Giovanni Triolo
- b Division of Rheumatology, Department of Internal Medicine , University of Palermo , Palermo , Italy
| | - Paola Cipriani
- a Division of Rheumatology, Department of Biotechnological and Applied Clinical Science , School of Medicine, University of L'Aquila , L'Aquila , Italy
| |
Collapse
|
73
|
The Wnt/β-catenin signaling/Id2 cascade mediates the effects of hypoxia on the hierarchy of colorectal-cancer stem cells. Sci Rep 2016; 6:22966. [PMID: 26965643 PMCID: PMC4786801 DOI: 10.1038/srep22966] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 02/22/2016] [Indexed: 01/03/2023] Open
Abstract
Hypoxia, a feature common to most solid tumors, is known to regulate many aspects of tumorigenesis. Recently, it was suggested that hypoxia increased the size of the cancer stem-cell (CSC) subpopulations and promoted the acquisition of a CSC-like phenotype. However, candidate hypoxia-regulated mediators specifically relevant to the stemness-related functions of colorectal CSCs have not been examined in detail. In the present study, we showed that hypoxia specifically promoted the self-renewal potential of CSCs. Through various in vitro studies, we found that hypoxia-induced Wnt/β-catenin signaling increased the occurrence of CSC-like phenotypes and the level of Id2 expression in colorectal-cancer cells. Importantly, the levels of hypoxia-induced CSC-sphere formation and Id2 expression were successfully attenuated by treatment with a Wnt/β-catenin-signaling inhibitor. We further demonstrated, for the first time, that the degree of hypoxia-induced CSC-sphere formation (CD44(+) subpopulation) in vitro and of tumor metastasis/dissemination in vivo were markedly suppressed by knocking down Id2 expression. Taken together, these data suggested that Wnt/β-catenin signaling mediated the hypoxia-induced self-renewal potential of colorectal-cancer CSCs through reactivating Id2 expression.
Collapse
|
74
|
Bakhashab S, Ahmed FW, Schulten HJ, Bashir A, Karim S, Al-Malki AL, Gari MA, Abuzenadah AM, Chaudhary AG, Alqahtani MH, Lary S, Ahmed F, Weaver JU. Metformin improves the angiogenic potential of human CD34⁺ cells co-incident with downregulating CXCL10 and TIMP1 gene expression and increasing VEGFA under hyperglycemia and hypoxia within a therapeutic window for myocardial infarction. Cardiovasc Diabetol 2016; 15:27. [PMID: 26861446 PMCID: PMC4748498 DOI: 10.1186/s12933-016-0344-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/26/2016] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in patients with diabetes mellitus (DM). To identify the most effective treatment for CVD, it is paramount to understand the mechanism behind cardioprotective therapies. Although metformin has been shown to reduce CVD in Type-2 DM clinical trials, the underlying mechanism remains unexplored. CD34(+) cell-based therapies offer a new treatment approach to CVD. The aim of this study was to investigate the effect of metformin on the angiogenic properties of CD34(+) cells under conditions mimicking acute myocardial infarction in diabetes. METHODS CD34(+) cells were cultured in 5.5 or 16.5 mmol/L glucose ± 0.01 mmol/L metformin and then additionally ± 4 % hypoxia. The paracrine function of CD34(+) cell-derived conditioned medium was assessed by measuring pro-inflammatory cytokines, vascular endothelial growth factor A (VEGFA), and using an in vitro tube formation assay for angiogenesis. Also, mRNA of CD34(+) cells was assayed by microarray and genes of interest were validated by qRT-PCR. RESULTS Metformin increased in vitro angiogenesis under hyperglycemia-hypoxia and augmented the expression of VEGFA. It also reduced the angiogenic-inhibitors, chemokine (C-X-C motif) ligand 10 (CXCL10) and tissue inhibitor of metalloproteinase 1 (TIMP1) mRNAs, which were upregulated under hyperglycemia-hypoxia. In addition metformin, increased expression of STEAP family member 4 (STEAP4) under euglycemia, indicating an anti-inflammatory effect. CONCLUSIONS Metformin has a dual effect by simultaneously increasing VEGFA and reducing CXCL10 and TIMP1 in CD34(+) cells in a model of the diabetic state combined with hypoxia. Therefore, these angiogenic inhibitors are promising therapeutic targets for CVD in diabetic patients. Moreover, our data are commensurate with a vascular protective effect of metformin and add to the understanding of underlying mechanisms.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia.
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Fahad W Ahmed
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
- Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne, UK.
| | - Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ayat Bashir
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | | | - Mamdooh A Gari
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Adel M Abuzenadah
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Adeel G Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mohammed H Alqahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Sahira Lary
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Farid Ahmed
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Jolanta U Weaver
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
- Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne, UK.
| |
Collapse
|
75
|
Liu J, Geng X, Li Y. Milky spots: omental functional units and hotbeds for peritoneal cancer metastasis. Tumour Biol 2016; 37:5715-26. [PMID: 26831659 PMCID: PMC4875158 DOI: 10.1007/s13277-016-4887-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/19/2016] [Indexed: 02/06/2023] Open
Abstract
As the most common metastatic disease of abdomen pelvic cavity cancer, peritoneal carcinomatosis (PC) renders significant negative impact on patient survival and quality of life. Invasive peritoneal exfoliated cancer cells (PECCs) preferentially select the omentum as a predominant target site for cancer cell colonization and proliferation compared with other tissues in the abdominal cavity. The precise pathogenic mechanism remains to be determined. As omental milky spots (MSs) are the major implantation site for malignant cells in peritoneal dissemination, researches on mechanisms of PC have been mainly focused on MS, primitive lymphoid tissues with unique structural features, and functional characteristics. To date, extensive biophysical and biochemical methods have been manipulated to investigate the MS exact function in the peritoneal cavity. This review summarized MS as hotbeds for PECC. The anatomical distribution was briefly described first. Then, MS histology was systematically reviewed, including morphological features, cellular constituents, and histological staining methods. At last, the roles of MS in PC pathological process were summarized with special emphasis on the distinct roles of macrophages.
Collapse
Affiliation(s)
- Jiuyang Liu
- Department of Oncology, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Xiafei Geng
- Department of Oncology, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Yan Li
- Department of Oncology, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China. .,Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital Affiliated to the Capital Medical University, Tieyilu 10, Yangfangdian, Haidian District, Beijing, 100038, People's Republic of China.
| |
Collapse
|
76
|
Biswas SK. Metabolic Reprogramming of Immune Cells in Cancer Progression. Immunity 2016; 43:435-49. [PMID: 26377897 DOI: 10.1016/j.immuni.2015.09.001] [Citation(s) in RCA: 495] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/25/2015] [Accepted: 08/31/2015] [Indexed: 11/25/2022]
Abstract
Immune cells play a key role in host defense against infection and cancer. Upon encountering danger signals, these cells undergo activation leading to a modulation in their immune functions. However, recent studies reveal that immune cells upon activation also show distinct metabolic changes that impact their immune functions. Such metabolic reprogramming and its functional effects are well known for cancer cells. Given that immune cells have emerged as crucial players in cancer progression, it is important to understand whether immune cells also undergo metabolic reprogramming in tumors and how this might affect their contribution in cancer progression. This emerging aspect of tumor-associated immune cells is reviewed here, discussing metabolic reprogramming of different immune cell types, the key pathways involved, and its impact on tumor progression.
Collapse
Affiliation(s)
- Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology & Research (A(∗)STAR), #04-06 Immunos, 8A Biomedical Grove, Singapore 138648, Singapore.
| |
Collapse
|
77
|
Huang WC, Chiang WH, Cheng YH, Lin WC, Yu CF, Yen CY, Yeh CK, Chern CS, Chiang CS, Chiu HC. Tumortropic monocyte-mediated delivery of echogenic polymer bubbles and therapeutic vesicles for chemotherapy of tumor hypoxia. Biomaterials 2015; 71:71-83. [DOI: 10.1016/j.biomaterials.2015.08.033] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/14/2015] [Accepted: 08/16/2015] [Indexed: 10/23/2022]
|
78
|
Jeong SK, Kim JS, Lee CG, Park YS, Kim SD, Yoon SO, Han DH, Lee KY, Jeong MH, Jo WS. Tumor associated macrophages provide the survival resistance of tumor cells to hypoxic microenvironmental condition through IL-6 receptor-mediated signals. Immunobiology 2015; 222:55-65. [PMID: 26705936 DOI: 10.1016/j.imbio.2015.11.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 10/23/2015] [Accepted: 11/22/2015] [Indexed: 01/05/2023]
Abstract
Hypoxia and infiltration of tumor-associated macrophages (TAM) are intrinsic features of the tumor microenvironment. Tumor cells that remain viable in hypoxic conditions often possess an increased survival potential and tend to grow aggressively. TAM also respond to a variety of signals in the hypoxic tumor microenvironment and express a more M2-like phenotype. In this study, the established mouse tumor tissues showed a dense infiltration of CD206+ macrophages at the junctions between the normoxic and hypoxic regions and an increased IL-6 receptor (IL-6R) expression of tumor cells in the areas of CD206+ TAM accumulation, which indicates a role of M2 phenotype TAM in survival adaptation of tumor cells preparing for an impending hypoxic injury before changes in oxygen availability. Cocultured mouse FM3A or human MCF-7 tumor cells with tumor infiltrating macrophages isolated from mouse tumor tissues and M2-polarized macrophages generated from human THP-1 cells, respectively, showed significantly decreased rate of cell death in cultures exposed to hypoxia. The acquisition of survival resistance was attributed to increased IL-6 production by M2 TAM and increased expression of IL-6R in tumor cells in the coculture system. MCF-7 cells cocultured with M2 TAM showed activated JAK1/STAT3 and Raf/MEK/JNK pathways contributing to tyrosine and serine phophorylation of STAT3, respectively. However, only tyrosine phosphorylated STAT3 was detected in the nucleus, which induced upregulation of Bcl-2 and downregulation of Bax and Bak. Finally, knockdown of IL-6R by small interfering RNA significantly counteracted coculture-induced signals and completely abolished the survival resistance to hypoxic injury. Thus, we present evidence for the role of M2 phenotype TAM in IL-6 receptor-mediated signals, particularly tyrosine phosphorylation of STAT3, responsible for the prosurvival adaptation of tumor cells to hypoxia.
Collapse
Affiliation(s)
- Soo Kyung Jeong
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea; Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea
| | - Joong Sun Kim
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - Chang Geun Lee
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - You-Soo Park
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - Sung Dae Kim
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - Sun Ok Yoon
- Transplantation Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Dong Hoon Han
- Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea
| | - Kyu Yeol Lee
- Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea
| | - Min Ho Jeong
- Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea.
| | - Wol Soon Jo
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea.
| |
Collapse
|
79
|
D'Ignazio L, Bandarra D, Rocha S. NF-κB and HIF crosstalk in immune responses. FEBS J 2015; 283:413-24. [PMID: 26513405 PMCID: PMC4864946 DOI: 10.1111/febs.13578] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022]
Abstract
Hypoxia and inflammation have been associated with a number of pathological conditions, in particular inflammatory diseases. While hypoxia is mainly associated with the activation of hypoxia‐inducible factors (HIFs), inflammation activates the family of transcription factor called nuclear factor‐kappa B (NF‐κB). An extensive crosstalk between these two main molecular players involved in hypoxia and inflammation has been demonstrated. This crosstalk includes common activating stimuli, shared regulators and targets. In this review, we discuss the current understanding of the role of NF‐κB and HIF in the context of the immune response. We review the crosstalk between HIF and NF‐κB in the control of the immune response in different immune cell types including macrophages, neutrophils and B and T cells. Furthermore the importance of the molecular crosstalk between HIFs and NF‐κB for a variety of medical conditions will be discussed.
Collapse
Affiliation(s)
- Laura D'Ignazio
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, UK
| | - Daniel Bandarra
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, UK
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, UK
| |
Collapse
|
80
|
Intense FDG uptake in an intra-articular localized giant-cell tumor of the tendon sheath (pigmented villonodular synovitis) mimics metastatic melanoma. Radiol Case Rep 2015; 4:343. [PMID: 27307839 PMCID: PMC4898176 DOI: 10.2484/rcr.v4i4.343] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We describe a patient with metastatic melanoma, one year following a clinical trial of VEGF
Collapse
|
81
|
Bone marrow-derived macrophages exclusively expressed caveolin-2: The role of inflammatory activators and hypoxia. Immunobiology 2015. [DOI: 10.1016/j.imbio.2015.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
82
|
Norton SE, Ward-Hartstonge KA, Taylor ES, Kemp RA. Immune cell interplay in colorectal cancer prognosis. World J Gastrointest Oncol 2015; 7:221-32. [PMID: 26483876 PMCID: PMC4606176 DOI: 10.4251/wjgo.v7.i10.221] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/12/2015] [Accepted: 08/25/2015] [Indexed: 02/05/2023] Open
Abstract
The immune response to colorectal cancer has proven to be a reliable measure of patient outcome in several studies. However, the complexity of the immune response in this disease is not well understood, particularly the interactions between tumour-associated cells and cells of the innate and adaptive immune system. This review will discuss the relationship between cancer associated fibroblasts and macrophages, as well as between macrophages and T cells, and demonstrate how each population may support or prevent tumour growth in a different immune environment.
Collapse
|
83
|
Noman MZ, Hasmim M, Messai Y, Terry S, Kieda C, Janji B, Chouaib S. Hypoxia: a key player in antitumor immune response. A Review in the Theme: Cellular Responses to Hypoxia. Am J Physiol Cell Physiol 2015; 309:C569-79. [PMID: 26310815 DOI: 10.1152/ajpcell.00207.2015] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The tumor microenvironment is a complex system, playing an important role in tumor development and progression. Besides cellular stromal components, extracellular matrix fibers, cytokines, and other metabolic mediators are also involved. In this review we outline the potential role of hypoxia, a major feature of most solid tumors, within the tumor microenvironment and how it contributes to immune resistance and immune suppression/tolerance and can be detrimental to antitumor effector cell functions. We also outline how hypoxic stress influences immunosuppressive pathways involving macrophages, myeloid-derived suppressor cells, T regulatory cells, and immune checkpoints and how it may confer tumor resistance. Finally, we discuss how microenvironmental hypoxia poses both obstacles and opportunities for new therapeutic immune interventions.
Collapse
Affiliation(s)
- Muhammad Zaeem Noman
- Institut National de la Santé et de la Recherche Médicale U1186, Equipe Labellisée Par La Ligue Contre Le Cancer, Gustave Roussy Campus, Villejuif, France
| | - Meriem Hasmim
- Institut National de la Santé et de la Recherche Médicale U1186, Equipe Labellisée Par La Ligue Contre Le Cancer, Gustave Roussy Campus, Villejuif, France
| | - Yosra Messai
- Institut National de la Santé et de la Recherche Médicale U1186, Equipe Labellisée Par La Ligue Contre Le Cancer, Gustave Roussy Campus, Villejuif, France
| | - Stéphane Terry
- Institut National de la Santé et de la Recherche Médicale U1186, Equipe Labellisée Par La Ligue Contre Le Cancer, Gustave Roussy Campus, Villejuif, France
| | - Claudine Kieda
- Centre for Molecular Biophysics, Cell Recognition, and Glycobiology, UPR 4301 Centre National de la Recherche Scientifique, Orléans, France; and
| | - Bassam Janji
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Salem Chouaib
- Institut National de la Santé et de la Recherche Médicale U1186, Equipe Labellisée Par La Ligue Contre Le Cancer, Gustave Roussy Campus, Villejuif, France;
| |
Collapse
|
84
|
Directing cell therapy to anatomic target sites in vivo with magnetic resonance targeting. Nat Commun 2015; 6:8009. [PMID: 26284300 PMCID: PMC4568295 DOI: 10.1038/ncomms9009] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 07/08/2015] [Indexed: 01/17/2023] Open
Abstract
Cell-based therapy exploits modified human cells to treat diseases but its targeted application in specific tissues, particularly those lying deep in the body where direct injection is not possible, has been problematic. Here we use a magnetic resonance imaging (MRI) system to direct macrophages carrying an oncolytic virus, Seprehvir, into primary and metastatic tumour sites in mice. To achieve this, we magnetically label macrophages with super-paramagnetic iron oxide nanoparticles and apply pulsed magnetic field gradients in the direction of the tumour sites. Magnetic resonance targeting guides macrophages from the bloodstream into tumours, resulting in increased tumour macrophage infiltration and reduction in tumour burden and metastasis. Our study indicates that clinical MRI scanners can not only track the location of magnetically labelled cells but also have the potential to steer them into one or more target tissues. Cell therapy requires the targeting of cells to specific sites in the body. Here Muthana et al. use a standard MRI scanner to direct oncolytic macrophages, labelled with magnetic nanoparticles, to primary and metastatic tumour sites in mice, and demonstrate that this leads to reduced tumour growth.
Collapse
|
85
|
Mechanisms of Hypoxic Up-Regulation of Versican Gene Expression in Macrophages. PLoS One 2015; 10:e0125799. [PMID: 26057378 PMCID: PMC4461269 DOI: 10.1371/journal.pone.0125799] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 03/26/2015] [Indexed: 12/22/2022] Open
Abstract
Hypoxia is a hallmark of many pathological tissues. Macrophages accumulate in hypoxic sites and up-regulate a range of hypoxia-inducible genes. The matrix proteoglycan versican has been identified as one such gene, but the mechanisms responsible for hypoxic induction are not fully characterised. Here we investigate the up-regulation of versican by hypoxia in primary human monocyte-derived macrophages (HMDM), and, intriguingly, show that versican mRNA is up-regulated much more highly (>600 fold) by long term hypoxia (5 days) than by 1 day of hypoxia (48 fold). We report that versican mRNA decay rates are not affected by hypoxia, demonstrating that hypoxic induction of versican mRNA is mediated by increased transcription. Deletion analysis of the promoter identified two regions required for high level promoter activity of luciferase reporter constructs in human macrophages. The hypoxia-inducible transcription factor HIF-1 has previously been implicated as a key potential regulator of versican expression in hypoxia, however our data suggest that HIF-1 up-regulation is unlikely to be principally responsible for the high levels of induction observed in HMDM. Treatment of HMDM with two distinct specific inhibitors of Phosphoinositide 3-kinase (PI3K), LY290042 and wortmannin, significantly reduced induction of versican mRNA by hypoxia and provides evidence of a role for PI3K in hypoxic up-regulation of versican expression.
Collapse
|
86
|
Leblond MM, Gérault AN, Corroyer-Dulmont A, MacKenzie ET, Petit E, Bernaudin M, Valable S. Hypoxia induces macrophage polarization and re-education toward an M2 phenotype in U87 and U251 glioblastoma models. Oncoimmunology 2015; 5:e1056442. [PMID: 26942063 PMCID: PMC4760330 DOI: 10.1080/2162402x.2015.1056442] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 11/28/2022] Open
Abstract
Hypoxia is a common feature of solid tumors, particularly in glioblastoma (GBM), and known to be a poor prognosis factor in GBM patients. The growth of GBM is also associated with a marked inflammation partially characterized by an accumulation of macrophage (MΦ) of the M2 phenotype. However, the transition between M1 MΦ (antitumoral) and M2 MΦ (protumoral) phenotypes is a dynamic process. We made the assumption that oxygen (O2) availability could be a major regulator of this transition and that the intratumoral O2 gradient is of importance. We evaluated, in vivo, the impact of hypoxia on MΦ tropism and polarization in two models of human GBM, well differentiated by their degree of hypoxia. MΦ migration in the tumor was more pronounced in the more hypoxic tumor of the two GBM models. In the more hypoxic of the models, we have shown that MΦ migrated at the tumor site only when hypoxia takes place. We also demonstrated that the acquisition of the M2 phenotype was clearly an evolving phenomenon with hypoxia as the major trigger for this transition. In support of these in vivo finding, M0 but also M1 MΦ cultured in moderate or severe hypoxia displayed a phenotype close to that of M2 MΦ whose phenotype was further reinforced by severe hypoxia. These results highlight the role of hypoxia in the aggressiveness of GBM, in part, by transforming MΦ such that a protumoral activity is expressed.
Collapse
Affiliation(s)
- Marine M Leblond
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Aurélie N Gérault
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Aurélien Corroyer-Dulmont
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Eric T MacKenzie
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Edwige Petit
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Myriam Bernaudin
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Samuel Valable
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| |
Collapse
|
87
|
Downs CA, Faulkner MS. Toxic stress, inflammation and symptomatology of chronic complications in diabetes. World J Diabetes 2015; 6:554-565. [PMID: 25987953 PMCID: PMC4434076 DOI: 10.4239/wjd.v6.i4.554] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/30/2014] [Accepted: 02/12/2015] [Indexed: 02/05/2023] Open
Abstract
Diabetes affects at least 382 million people worldwide and the incidence is expected to reach 592 million by 2035. The incidence of diabetes in youth is skyrocketing as evidenced by a 21% increase in type 1 diabetes and a 30.5% increase in type 2 diabetes in the United States between 2001 and 2009. The effects of toxic stress, the culmination of biological and environmental interactions, on the development of diabetes complications is gaining attention. Stress impacts the hypothalamus-pituitary-adrenal axis and contributes to inflammation, a key biological contributor to the pathogenesis of diabetes and its associated complications. This review provides an overview of common diabetic complications such as neuropathy, cognitive decline, depression, nephropathy and cardiovascular disease. The review also provides a discussion of the role of inflammation and stress in the development and progression of chronic complications of diabetes, associated symptomatology and importance of early identification of symptoms of depression, fatigue, exercise intolerance and pain.
Collapse
|
88
|
Matsuura Y, Yamashita A, Iwakiri T, Sugita C, Okuyama N, Kitamura K, Asada Y. Vascular wall hypoxia promotes arterial thrombus formation via augmentation of vascular thrombogenicity. Thromb Haemost 2015; 114:158-72. [PMID: 25833755 DOI: 10.1160/th14-09-0794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/30/2015] [Indexed: 12/15/2022]
Abstract
Atherosclerotic lesions represent a hypoxic milieu. However, the significance of this milieu in atherothrombosis has not been established. We aimed to assess the hypothesis that vascular wall hypoxia promotes arterial thrombus formation. We examined the relation between vascular wall hypoxia and arterial thrombus formation using a rabbit model in which arterial thrombosis was induced by 0.5 %-cholesterol diet and repeated balloon injury of femoral arteries. Vascular wall hypoxia was immunohistochemically detected by pimonidazole hydrochloride, a hypoxia marker. Rabbit neointima and THP-1 macrophages were cultured to analyse prothrombotic factor expression under hypoxic conditions (1 % O2). Prothrombotic factor expression and nuclear localisation of hypoxia-inducible factor (HIF)-1α and nuclear factor-kappa B (NF-κB) p65 were immunohistochemically assessed using human coronary atherectomy plaques. Hypoxic areas were localised in the macrophage-rich deep portion of rabbit neointima and positively correlated with the number of nuclei immunopositive for HIF-1α and NF-κB p65, and tissue factor (TF) expression. Immunopositive areas for glycoprotein IIb/IIIa and fibrin in thrombi were significantly correlated with hypoxic areas in arteries. TF and plasminogen activator inhibitor-1 (PAI-1) expression was increased in neointimal tissues and/or macrophages cultured under hypoxia, and both were suppressed by inhibitors of either HIF-1 or NF-κB. In human coronary plaques, the number of HIF-1α-immunopositive nuclei was positively correlated with that of NF-κB-immunopositive nuclei and TF-immunopositive and PAI-1-immunopositive area, and it was significantly higher in thrombotic plaques. Vascular wall hypoxia augments the thrombogenic potential of atherosclerotic plaque and thrombus formation on plaques via prothrombotic factor upregulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yujiro Asada
- Yujiro Asada, MD, Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan, Tel.: + 81 985 85 2810, Fax: + 81 985 85 7614, E-mail:
| |
Collapse
|
89
|
Newby AC. Metalloproteinases promote plaque rupture and myocardial infarction: A persuasive concept waiting for clinical translation. Matrix Biol 2015; 44-46:157-66. [PMID: 25636537 DOI: 10.1016/j.matbio.2015.01.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/20/2015] [Accepted: 01/20/2015] [Indexed: 02/05/2023]
Abstract
Atherosclerotic plaque rupture provokes most myocardial infarctions. Matrix metalloproteinases (MMPs) have counteracting roles in intimal thickening, which stabilizes plaques, on the one hand and extracellular matrix destruction that leads to plaque rupture on the other. This review briefly summarizes the key points supporting the involvement of individual MMPs in provoking plaque rupture and discusses the barriers that stand in the way of clinical translation, which can be itemised as follows: structural and functional complexity of the MMP family; lack of adequate preclinical models partly owing to different expression patterns of MMPs and TIMPs in mouse and human macrophages; the need to target individual MMPs selectively; the difficulties in establishing causality in human studies; and the requirement for surrogate markers of efficacy. Overcoming these barriers would open the way to new treatments that could have a major impact on cardiovascular mortality worldwide.
Collapse
Affiliation(s)
- Andrew C Newby
- University of Bristol, School of Clinical Sciences and Bristol Heart Institute, Bristol, UK.
| |
Collapse
|
90
|
Muraille E, Leo O, Moser M. TH1/TH2 paradigm extended: macrophage polarization as an unappreciated pathogen-driven escape mechanism? Front Immunol 2014; 5:603. [PMID: 25505468 PMCID: PMC4244692 DOI: 10.3389/fimmu.2014.00603] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/10/2014] [Indexed: 02/04/2023] Open
Abstract
The classical view of the Th1/Th2 paradigm posits that the pathogen nature, infectious cycle, and persistence represent key parameters controlling the choice of effector mechanisms operating during an immune response. Thus, efficient Th1 responses are triggered by replicating intracellular pathogens, while Th2 responses would control helminth infection and promote tissue repair during the resolution phase of an infectious event. However, this vision does not account for a growing body of data describing how pathogens exploit the polarization of the host immune response to their own benefit. Recently, the study of macrophages has illustrated a novel aspect of this arm race between pathogens and the immune system, and the central role of macrophages in homeostasis, repair and defense of all tissues is now fully appreciated. Like T lymphocytes, macrophages differentiate into distinct effectors including classically (M1) and alternatively (M2) activated macrophages. Interestingly, in addition to represent immune effectors, M1/M2 cells have been shown to represent potential reservoir cells to a wide range of intracellular pathogens. Subversion of macrophage cell metabolism by microbes appears as a recently uncovered immune escape strategy. Upon infection, several microbial agents have been shown to activate host metabolic pathways leading to the production of nutrients necessary to their long-term persistence in host. The purpose of this review is to summarize and discuss the strategies employed by pathogens to manipulate macrophage differentiation, and in particular their basic cell metabolism, to favor their own growth while avoiding immune control.
Collapse
Affiliation(s)
- Eric Muraille
- Laboratory of Parasitology, Faculty of Medicine, Université Libre de Bruxelles , Brussels , Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Faculty of Sciences, Université Libre de Bruxelles , Gosselies , Belgium
| | - Muriel Moser
- Laboratory of Immunobiology, Faculty of Sciences, Université Libre de Bruxelles , Gosselies , Belgium
| |
Collapse
|
91
|
Tazzyman S, Murdoch C, Yeomans J, Harrison J, Muthana M. Macrophage-mediated response to hypoxia in disease. HYPOXIA 2014; 2:185-196. [PMID: 27774476 PMCID: PMC5045066 DOI: 10.2147/hp.s49717] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hypoxia plays a critical role in the pathobiology of various inflamed, diseased tissues, including malignant tumors, atherosclerotic plaques, myocardial infarcts, the synovia of rheumatoid arthritic joints, healing wounds, and sites of bacterial infection. These areas of hypoxia form when the blood supply is occluded and/or the oxygen supply is unable to keep pace with cell growth and/or infiltration of inflammatory cells. Macrophages are ubiquitous in all tissues of the body and exhibit great plasticity, allowing them to perform divergent functions, including, among others, patrolling tissue, combating invading pathogens and tumor cells, orchestrating wound healing, and restoring homeostasis after an inflammatory response. The number of tissue macrophages increases markedly with the onset and progression of many pathological states, with many macrophages accumulating in avascular and necrotic areas, where they are exposed to hypoxia. Recent studies show that these highly versatile cells then respond rapidly to the hypoxia present by altering their expression of a wide array of genes. Here we review the evidence for hypoxia-driven macrophage inflammatory responses in various disease states, and how this influences disease progression and treatment.
Collapse
Affiliation(s)
| | | | | | | | - Munitta Muthana
- Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| |
Collapse
|
92
|
Nonsurgical giant cell tumour of the tendon sheath or of the diffuse type: are MRI or 18F-FDG PET/CT able to provide an accurate prediction of long-term outcome? Eur J Nucl Med Mol Imaging 2014; 42:397-408. [PMID: 25367748 DOI: 10.1007/s00259-014-2938-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/08/2014] [Indexed: 01/18/2023]
Abstract
PURPOSE To investigate whether MRI (RECIST 1.1, WHO criteria and the volumetric approach) or (18)F-FDG PET/CT (PERCIST 1.0) are able to predict long-term outcome in nonsurgical patients with giant cell tumour of the tendon sheath or of the diffuse type (GCT-TS/DT). METHODS Fifteen "nonsurgical" patients with a histological diagnosis of GCT-TS/DT were divided into two groups: symptomatic patients receiving targeted therapy and asymptomatic untreated patients. All 15 patients were evaluated by MRI of whom 10 were treated, and a subgroup of 7 patients were evaluated by PET/CT of whom 4 were treated. Early evolution was assessed according to MRI and PET/CT scans at baseline and during follow-up. Cohen's kappa coefficient was used to evaluate the degree of agreement between PERCIST 1.0, RECIST 1.1, WHO criteria, volumetric approaches and the reference standard (long-term outcome, delay 505 ± 457 days). The response rate in symptomatic patients with GCT-TS/DT receiving targeted therapy was also assessed in a larger population that included additional patients obtained from a review of the literature. RESULTS The kappa coefficients for agreement between RECIST/WHO/volumetric criteria and outcome (15 patients) were respectively: 0.35 (p = 0.06), 0.26 (p = 0.17) and 0.26 (p = 0.17). In the PET/CT subgroup (7 patients), PERCIST was in perfect agreement with the late symptomatic evolution (kappa = 1, p < 0.05). In the treated symptomatic group including the additional patients from the literature the response rates to targeted therapies according to late symptomatic assessment, and PERCIST and RECIST criteria were: 65 % (22/34), 77 % (10/13) and 26 % (10/39). CONCLUSION (18)F-FDG PET/CT with PERCIST is a promising approach to the prediction of the long-term outcome in GCT-TS/DT and may avoid unnecessary treatments, toxicity and costs. On MRI, WHO and volumetric approaches are not more effective than RECIST using the current thresholds.
Collapse
|
93
|
Sotoodehnejadnematalahi F, Burke B. Human activated macrophages and hypoxia: a comprehensive review of the literature. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2014; 17:820-30. [PMID: 25691922 PMCID: PMC4328089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 07/01/2014] [Indexed: 10/27/2022]
Abstract
Macrophages accumulate in poorly vascularised and hypoxic sites including solid tumours, wounds and sites of infection and inflammation where they can be exposed to low levels of oxygen for long periods. Up to date, different studies have shown that a number of transcription factors are activated by hypoxia which in turn activate a broad array of mitogenic, pro-invasive, pro-angiogenic, and pro-metastatic genes. On the other hand, macrophages respond to hypoxia by up-regulating several genes which are chief factors in angiogenesis and tumorigenesis. Therefore, in this review article we focus mainly on the role of macrophages during inflammation and discuss their response to hypoxia by regulating a diverse array of transcription factors. We also review the existing literatures on hypoxia and its cellular and molecular mechanism which mediates macrophages activation.
Collapse
Affiliation(s)
- Fattah Sotoodehnejadnematalahi
- Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran,Corresponding author: Fattah Sotoodehnejadnematalahi. Department of Stem Cells and Developmental Biology, Royan Institute, Banihashem Sq., Banihashem St., Resalat highway, Tehran, Iran. Royan Regenerative Medicine and Cell Therapy Centre. Tel: +98-21-22306485; Fax: +98-21-22310406;
| | - Bernard Burke
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| |
Collapse
|
94
|
Marsch E, Theelen TL, Demandt JAF, Jeurissen M, van Gink M, Verjans R, Janssen A, Cleutjens JP, Meex SJR, Donners MM, Haenen GR, Schalkwijk CG, Dubois LJ, Lambin P, Mallat Z, Gijbels MJ, Heemskerk JWM, Fisher EA, Biessen EAL, Janssen BJ, Daemen MJAP, Sluimer JC. Reversal of hypoxia in murine atherosclerosis prevents necrotic core expansion by enhancing efferocytosis. Arterioscler Thromb Vasc Biol 2014; 34:2545-53. [PMID: 25256233 DOI: 10.1161/atvbaha.114.304023] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Advanced murine and human plaques are hypoxic, but it remains unclear whether plaque hypoxia is causally related to atherogenesis. Here, we test the hypothesis that reversal of hypoxia in atherosclerotic plaques by breathing hyperoxic carbogen gas will prevent atherosclerosis. APPROACH AND RESULTS Low-density lipoprotein receptor-deficient mice (LDLR(-/-)) were fed a Western-type diet, exposed to carbogen (95% O2, 5% CO2) or air, and the effect on plaque hypoxia, size, and phenotype was studied. First, the hypoxic marker pimonidazole was detected in murine LDLR(-/-) plaque macrophages from plaque initiation onwards. Second, the efficacy of breathing carbogen (90 minutes, single exposure) was studied. Compared with air, carbogen increased arterial blood pO2 5-fold in LDLR(-/-) mice and reduced plaque hypoxia in advanced plaques of the aortic root (-32%) and arch (-84%). Finally, the effect of repeated carbogen exposure on progression of atherosclerosis was studied in LDLR(-/-) mice fed a Western-type diet for an initial 4 weeks, followed by 4 weeks of diet and carbogen or air (both 90 min/d). Carbogen reduced plaque hypoxia (-40%), necrotic core size (-37%), and TUNEL(+) (terminal uridine nick-end labeling positive) apoptotic cell content (-50%) and increased efferocytosis of apoptotic cells by cluster of differentiation 107b(+) (CD107b, MAC3) macrophages (+36%) in advanced plaques of the aortic root. Plaque size, plasma cholesterol, hematopoiesis, and systemic inflammation were unchanged. In vitro, hypoxia hampered efferocytosis by bone marrow-derived macrophages, which was dependent on the receptor Mer tyrosine kinase. CONCLUSIONS Carbogen restored murine plaque oxygenation and prevented necrotic core expansion by enhancing efferocytosis, likely via Mer tyrosine kinase. Thus, plaque hypoxia is causally related to necrotic core expansion.
Collapse
Affiliation(s)
- Elke Marsch
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Thomas L Theelen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Jasper A F Demandt
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Mike Jeurissen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Mathijs van Gink
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Robin Verjans
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Anique Janssen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Jack P Cleutjens
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Steven J R Meex
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Marjo M Donners
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Guido R Haenen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Casper G Schalkwijk
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Ludwig J Dubois
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Philippe Lambin
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Ziad Mallat
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Marion J Gijbels
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Johan W M Heemskerk
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Edward A Fisher
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Erik A L Biessen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Ben J Janssen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Mat J A P Daemen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Judith C Sluimer
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.).
| |
Collapse
|
95
|
Folco EJ, Sukhova GK, Quillard T, Libby P. Moderate hypoxia potentiates interleukin-1β production in activated human macrophages. Circ Res 2014; 115:875-83. [PMID: 25185259 DOI: 10.1161/circresaha.115.304437] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Inflammation drives atherogenesis. Animal and human studies have implicated interleukin-1β (IL-1β) in this disease. Moderate hypoxia, a condition that prevails in the atherosclerotic plaque, may conspire with inflammation and contribute to the evolution and complications of atherosclerosis through mechanisms that remain incompletely understood. OBJECTIVE This study investigated the links between hypoxia and inflammation by testing the hypothesis that moderate hypoxia modulates IL-1β production in activated human macrophages. METHODS AND RESULTS Our results demonstrated that hypoxia enhances pro-IL-1β protein, but not mRNA, expression in lipopolysaccharide-stimulated human macrophages. We show that hypoxia limits the selective targeting of pro-IL-1β to autophagic degradation, thus prolonging its half-life and promoting its intracellular accumulation. Furthermore, hypoxia increased the expression of NLRP3, a limiting factor in NLRP3 inflammasome function, and augmented caspase-1 activation in lipopolysaccharide-primed macrophages. Consequently, hypoxic human macrophages secreted higher amounts of mature IL-1β than did normoxic macrophages after treatment with crystalline cholesterol, an endogenous danger signal that contributes to atherogenesis. In human atherosclerotic plaques, IL-1β localizes predominantly to macrophage-rich regions that express activated caspase-1 and the hypoxia markers hypoxia-inducible factor 1α and hexokinase-2, as assessed by immunohistochemical staining of carotid endarterectomy specimens. CONCLUSIONS These results indicate that hypoxia potentiates IL-1β expression in cultured human macrophages and in the context of atheromata, therefore unveiling a novel proinflammatory mechanism that may participate in atherogenesis.
Collapse
Affiliation(s)
- Eduardo J Folco
- From the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Galina K Sukhova
- From the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Thibaut Quillard
- From the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- From the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
96
|
Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel) 2014; 6:1670-90. [PMID: 25125485 PMCID: PMC4190561 DOI: 10.3390/cancers6031670] [Citation(s) in RCA: 1169] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/27/2014] [Accepted: 08/05/2014] [Indexed: 02/07/2023] Open
Abstract
During tumor progression, circulating monocytes and macrophages are actively recruited into tumors where they alter the tumor microenvironment to accelerate tumor progression. Macrophages shift their functional phenotypes in response to various microenvironmental signals generated from tumor and stromal cells. Based on their function, macrophages are divided broadly into two categories: classical M1 and alternative M2 macrophages. The M1 macrophage is involved in the inflammatory response, pathogen clearance, and antitumor immunity. In contrast, the M2 macrophage influences an anti-inflammatory response, wound healing, and pro-tumorigenic properties. Tumor-associated macrophages (TAMs) closely resemble the M2-polarized macrophages and are critical modulators of the tumor microenvironment. Clinicopathological studies have suggested that TAM accumulation in tumors correlates with a poor clinical outcome. Consistent with that evidence, experimental and animal studies have supported the notion that TAMs can provide a favorable microenvironment to promote tumor development and progression. In this review article, we present an overview of mechanisms responsible for TAM recruitment and highlight the roles of TAMs in the regulation of tumor angiogenesis, invasion, metastasis, immunosuppression, and chemotherapeutic resistance. Finally, we discuss TAM-targeting therapy as a promising novel strategy for an indirect cancer therapy.
Collapse
|
97
|
Abstract
Cellular hypoxic preconditioning is being employed to obtain complex, yet physiological, secretomes rich is angiogenic factors. We previously proposed exposing peripheral blood cells (PBCs) to hypoxic stress stimulation, and demonstrated that controlled release of PBC-derived factor mixtures induces directional microvessel growth in vitro. Hypoxia therefore provides a useful tool for enhancing the angiogenic potential of blood plasma, by generating compositions based on PBCs' natural responses to a wound-like microenvironment. Here, we discuss various methods for preparing and delivering Hypoxia Preconditioned Plasma (HPP), i.e., plasma derived after extracorporeal conditioning of anticoagulated blood under physiological temperature and hypoxia. Special emphasis is given to those approaches that will likely facilitate the clinical translation of HPP-based therapies. We finally draw a comparison between HPP and other, currently available blood-based products, and present the case that its arrival paves the way for developing next-generation autologous therapies toward angiogenesis-supported tissue repair and regeneration.
Collapse
Affiliation(s)
- Ektoras Hadjipanayi
- Experimental Plastic Surgery; Clinic for Plastic and Hand Surgery; Klinikum Rechts der Isar, Technische Universität München; Munich, Germany; Department of Plastic, Reconstructive, Hand and Burn Surgery; Bogenhausen Hospital; Munich, Germany
| | - Arndt F Schilling
- Experimental Plastic Surgery; Clinic for Plastic and Hand Surgery; Klinikum Rechts der Isar, Technische Universität München; Munich, Germany; Center for Applied New Technologies in Engineering for Regenerative Medicine (Canter); Munich, Germany
| |
Collapse
|
98
|
Singh A, Talekar M, Raikar A, Amiji M. Macrophage-targeted delivery systems for nucleic acid therapy of inflammatory diseases. J Control Release 2014; 190:515-30. [PMID: 24747762 DOI: 10.1016/j.jconrel.2014.04.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/21/2014] [Indexed: 01/10/2023]
Abstract
Inflammation is an immune response that marks several pathophysiological conditions in our body. Though adaptive immune cells play a major role in the progression of the disease, components of innate immune system, mainly monocytes and macrophages play the central role in onset of inflammation. Tissue-associated macrophages are widely distributed in the body showing tremendous anatomical and functional diversity and are actively involved in maintaining the homeostasis. They exhibit different phenotypes depending on their residing tissue microenvironment and the two major functional phenotypes are classically activated M1 phenotype showing pro-inflammatory characteristics and alternatively activated M2 phenotype demonstrating anti-inflammatory nature. Several cytokines, chemokines and other regulatory mediators delicately govern the balance of the two phenotypes in a tissue. This balance, however, is subverted during infection, injury or autoimmune response leading to increased population of M1 phenotype and subsequent chronic inflammatory disease states. This review underlines the role of macrophages in inflammatory diseases with an insight into potential molecular targets for nucleic acid therapy. Finally, some recent nanotechnology-based approaches to devise macrophage-specific targeted therapy have been highlighted.
Collapse
Affiliation(s)
- Amit Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA
| | - Meghna Talekar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA
| | - Ankita Raikar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston 02115, USA.
| |
Collapse
|
99
|
Stockmann C, Schadendorf D, Klose R, Helfrich I. The impact of the immune system on tumor: angiogenesis and vascular remodeling. Front Oncol 2014; 4:69. [PMID: 24782982 PMCID: PMC3986554 DOI: 10.3389/fonc.2014.00069] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/20/2014] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, as well as inflammation with massive infiltration of leukocytes are hallmarks of various tumor entities. Various epidemiological, clinical, and experimental studies have not only demonstrated a link between chronic inflammation and cancer onset but also shown that immune cells from the bone marrow such as tumor-infiltrating macrophages significantly influence tumor progression. Tumor angiogenesis is critical for tumor development as tumors have to establish a blood supply in order to progress. Although tumor cells were first believed to fuel tumor angiogenesis, numerous studies have shown that the tumor microenvironment and infiltrating immune cell subsets are important for regulating the process of tumor angiogenesis. These infiltrates involve the adaptive immune system including several types of lymphocytes as well as cells of the innate immunity such as macrophages, neutrophils, eosinophils, mast cells, dendritic cells, and natural killer cells. Besides their known immune function, these cells are now recognized for their crucial role in regulating the formation and the remodeling of blood vessels in the tumor. In this review, we will discuss for each cell type the mechanisms that regulate the vascular phenotype and its impact on tumor growth and metastasis.
Collapse
Affiliation(s)
- Christian Stockmann
- UMR 970, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale (INSERM) , Paris , France
| | - Dirk Schadendorf
- Skin Cancer Unit, Dermatology Department, Medical Faculty, University Duisburg-Essen , Essen , Germany
| | - Ralph Klose
- UMR 970, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale (INSERM) , Paris , France
| | - Iris Helfrich
- Skin Cancer Unit, Dermatology Department, Medical Faculty, University Duisburg-Essen , Essen , Germany
| |
Collapse
|
100
|
Increased metabolites of 5-lipoxygenase from hypoxic ovarian cancer cells promote tumor-associated macrophage infiltration. Oncogene 2014; 34:1241-52. [PMID: 24662827 DOI: 10.1038/onc.2014.85] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/11/2014] [Accepted: 02/25/2014] [Indexed: 12/27/2022]
Abstract
5-lipoxygenase (5-LOX), a member of the lipoxygenase gene family, is a key enzyme assisting in the conversion of arachidonic acid to 5-HETE and leukotrienes. Tumor-associated macrophages (TAMs) have a critical role in the progression and metastasis of many tumors, including ovarian tumors. Moreover, TAMs are often found in a high density in the hypoxic areas of tumors. However, the relevant mechanisms have not been studied explicitly until now. In this study, we found that the expression of 5-LOX strongly correlated with the density of TAMs in hypoxic areas of human ovarian tumor tissues. In cultured ovarian cancer cells, 5-LOX metabolites were increased under hypoxic conditons. Increased 5-LOX metabolites from hypoxic ovarian cancer cells promoted migration and invasion of macrophages, which was further demonstrated to be mediated by the upregulation of matrix metalloproteinase (MMP)-7 expression through the p38 pathway. Besides, we also showed that 5-LOX metabolites enhanced the release of tumor necrosis factor (TNF-α) and heparin-binding epidermal growth factor-like growth factor through upregulation of MMP-7. Furthermore, in animal models, Zileuton (a selective and specific 5-LOX inhibitor) reduced the MMP-7 expression and the number of macrophages infiltrating in the xenograft. Our findings suggest for the first time that increased metabolites of 5-LOX from hypoxic ovarian cancer cells promote TAM infiltration. These results of this study have immediate translational implications for the therapeutic exploitation of TAMs.
Collapse
|