301
|
Chockley PJ, Keshamouni VG. Immunological Consequences of Epithelial-Mesenchymal Transition in Tumor Progression. THE JOURNAL OF IMMUNOLOGY 2017; 197:691-8. [PMID: 27431984 DOI: 10.4049/jimmunol.1600458] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/18/2016] [Indexed: 12/26/2022]
Abstract
Microenvironments that tumor cells encounter are different during the stages of cancer progression-primary tumor, metastasis, and at the metastatic site. This suggests potential differences in immune surveillance of primary tumor and metastasis. Epithelial-mesenchymal transition (EMT) is a key reversible process in which cancer cells transition into highly motile and invasive cells for dissemination. Only a tiny proportion successfully metastasize, supporting the notion of metastasis-specific immune surveillance. EMT involves extensive molecular reprogramming of cells conferring many clinically relevant features to cancer cells and affects tumor cell interactions within the tumor microenvironment. We review the impact of tumor immune infiltrates on tumor cell EMT and the consequences of EMT in shaping the immune microenvironment of tumors. The usefulness of EMT as a model to investigate metastasis-specific immune surveillance mechanisms are also explored. Finally, we discuss potential implications of EMT for tumor immunogenicity, as well as current immunotherapies and future strategies.
Collapse
Affiliation(s)
- Peter J Chockley
- Graduate Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI 48109; and
| | - Venkateshwar G Keshamouni
- Graduate Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI 48109; and Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| |
Collapse
|
302
|
Seager RJ, Hajal C, Spill F, Kamm RD, Zaman MH. Dynamic interplay between tumour, stroma and immune system can drive or prevent tumour progression. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017; 3. [PMID: 30079253 DOI: 10.1088/2057-1739/aa7e86] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the tumour microenvironment, cancer cells directly interact with both the immune system and the stroma. It is firmly established that the immune system, historically believed to be a major part of the body's defence against tumour progression, can be reprogrammed by tumour cells to be ineffective, inactivated, or even acquire tumour promoting phenotypes. Likewise, stromal cells and extracellular matrix can also have pro-and anti-tumour properties. However, there is strong evidence that the stroma and immune system also directly interact, therefore creating a tripartite interaction that exists between cancer cells, immune cells and tumour stroma. This interaction contributes to the maintenance of a chronically inflamed tumour microenvironment with pro-tumorigenic immune phenotypes and facilitated metastatic dissemination. A comprehensive understanding of cancer in the context of dynamical interactions of the immune system and the tumour stroma is therefore required to truly understand the progression toward and past malignancy.
Collapse
Affiliation(s)
- R J Seager
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston MA 02215
| | - Cynthia Hajal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Fabian Spill
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston MA 02215.,Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston MA 02215.,Howard Hughes Medical Institute, Boston University, Boston, MA 02215
| |
Collapse
|
303
|
Abstract
Myeloid cell recruitment to sites of infection and injury started out as a simple model that has been referred to as the universal concept of leukocyte recruitment. However, as we gain more insight into the different mechanisms, it is becoming clear that each organ and perhaps even each cell has its own unique mechanism of recruitment. Moreover, as the ability to visualize specific cell types in specific organs becomes more accessible, it is also becoming clear that there are resident populations of leukocytes, some within the tissues and others attached to the vasculature of tissues, the latter poised to affect the local environment. In this review, we will first highlight the imaging approaches that have allowed us to gain spectacular insight into locale and function of specific cell types, and then we will discuss what we have learned from this approach as far as myeloid cells are concerned. We will also highlight some of the gaps in our knowledge, which exist almost certainly because of the challenges of being able to visualize certain compartments of the body.
Collapse
|
304
|
Jung K, Heishi T, Khan OF, Kowalski PS, Incio J, Rahbari NN, Chung E, Clark JW, Willett CG, Luster AD, Yun SH, Langer R, Anderson DG, Padera TP, Jain RK, Fukumura D. Ly6Clo monocytes drive immunosuppression and confer resistance to anti-VEGFR2 cancer therapy. J Clin Invest 2017; 127:3039-3051. [PMID: 28691930 DOI: 10.1172/jci93182] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/19/2017] [Indexed: 12/13/2022] Open
Abstract
Current anti-VEGF therapies for colorectal cancer (CRC) provide limited survival benefit, as tumors rapidly develop resistance to these agents. Here, we have uncovered an immunosuppressive role for nonclassical Ly6Clo monocytes that mediates resistance to anti-VEGFR2 treatment. We found that the chemokine CX3CL1 was upregulated in both human and murine tumors following VEGF signaling blockade, resulting in recruitment of CX3CR1+Ly6Clo monocytes into the tumor. We also found that treatment with VEGFA reduced expression of CX3CL1 in endothelial cells in vitro. Intravital microscopy revealed that CX3CR1 is critical for Ly6Clo monocyte transmigration across the endothelium in murine CRC tumors. Moreover, Ly6Clo monocytes recruit Ly6G+ neutrophils via CXCL5 and produce IL-10, which inhibits adaptive immunity. Preventing Ly6Clo monocyte or Ly6G+ neutrophil infiltration into tumors enhanced inhibition of tumor growth with anti-VEGFR2 therapy. Furthermore, a gene therapy using a nanoparticle formulated with an siRNA against CX3CL1 reduced Ly6Clo monocyte recruitment and improved outcome of anti-VEGFR2 therapy in mouse CRCs. Our study unveils an immunosuppressive function of Ly6Clo monocytes that, to our knowledge, has yet to be reported in any context. We also reveal molecular mechanisms underlying antiangiogenic treatment resistance, suggesting potential immunomodulatory strategies to enhance the long-term clinical outcome of anti-VEGF therapies.
Collapse
Affiliation(s)
- Keehoon Jung
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Takahiro Heishi
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Omar F Khan
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
| | - Piotr S Kowalski
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
| | - Joao Incio
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nuh N Rahbari
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Euiheon Chung
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jeffrey W Clark
- Department of Hematology/Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Seok Hyun Yun
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
| | - Timothy P Padera
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dai Fukumura
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
305
|
Alvey C, Discher DE. Engineering macrophages to eat cancer: from "marker of self" CD47 and phagocytosis to differentiation. J Leukoc Biol 2017; 102:31-40. [PMID: 28522599 PMCID: PMC6608056 DOI: 10.1189/jlb.4ri1216-516r] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
The ability of a macrophage to engulf and break down invading cells and other targets provides a first line of immune defense in nearly all tissues. This defining ability to "phagos" or devour can subsequently activate the entire immune system against foreign and diseased cells, and progress is now being made on a decades-old idea of directing macrophages to phagocytose specific targets, such as cancer cells. Engineered T cells provide precedence with recent clinical successes against liquid tumors, but solid tumors remain a challenge, and a handful of clinical trials seek to exploit the abundance of tumor-associated macrophages instead. Although macrophage differentiation into such phenotypes with deficiencies in phagocytic ability can raise challenges, newly recognized features of cancer cells that might be manipulated to increase the phagocytosis of those cells include ≥1 membrane protein, CD47, which broadly inhibits phagocytosis and is abundantly expressed on all healthy cells. Physical properties of the target also influence phagocytosis and again relate-via cytoskeleton forces-to differentiation pathways in solid tumors. Such pathways extend to mechanosensing by the nuclear lamina, which is known to influence signaling by soluble retinoids that can regulate the macrophage SIRPα, the receptor for CD47. Here, we highlight some of those past, present, and rapidly emerging efforts to understand and control macrophages for cancer therapy.
Collapse
Affiliation(s)
- Cory Alvey
- Systems Pharmacology and Translational Therapeutics Graduate Group, Physical Sciences Oncology Center at Penn, Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dennis E Discher
- Systems Pharmacology and Translational Therapeutics Graduate Group, Physical Sciences Oncology Center at Penn, Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
306
|
Secklehner J, Lo Celso C, Carlin LM. Intravital microscopy in historic and contemporary immunology. Immunol Cell Biol 2017; 95:506-513. [PMID: 28366932 PMCID: PMC6095455 DOI: 10.1038/icb.2017.25] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 12/31/2022]
Abstract
In this review, we discuss intravital microscopy of immune cells, starting from its historic origins to current applications in diverse organs. It is clear from a quantitative review of the literature that intravital microscopy is a key tool in both historic and contemporary immunological research, providing unique advances in our understanding of immune responses. We have chosen to focus this review on how intravital microscopy methodologies are used to image specific organs or systems and we present recent descriptions of fundamental immunological processes that could not have been achieved by other methods. The following target organs/systems are discussed in more detail: cremaster muscle, skin (ear and dorsal skin fold chamber), lymph node, liver, lung, mesenteric vessels, carotid artery, bone marrow, brain, spleen, foetus and lastly vessels of the knee joint.
Collapse
Affiliation(s)
- Judith Secklehner
- Cancer Research UK Beatson Institute, Garscube Campus, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Inflammation, Repair & Development, National Heart & Lung Institute, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| | - Cristina Lo Celso
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
- The Francis Crick Institute, 1 Midland Road, London NW1A 1AT, UK
| | - Leo M. Carlin
- Cancer Research UK Beatson Institute, Garscube Campus, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Inflammation, Repair & Development, National Heart & Lung Institute, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| |
Collapse
|
307
|
Alvey CM, Spinler KR, Irianto J, Pfeifer CR, Hayes B, Xia Y, Cho S, Dingal PCPD, Hsu J, Smith L, Tewari M, Discher DE. SIRPA-Inhibited, Marrow-Derived Macrophages Engorge, Accumulate, and Differentiate in Antibody-Targeted Regression of Solid Tumors. Curr Biol 2017; 27:2065-2077.e6. [PMID: 28669759 DOI: 10.1016/j.cub.2017.06.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022]
Abstract
Marrow-derived macrophages are highly phagocytic, but whether they can also traffic into solid tumors and engulf cancer cells is questionable, given the well-known limitations of tumor-associated macrophages (TAMs). Here, SIRPα on macrophages from mouse and human marrow was inhibited to block recognition of its ligand, the "marker of self" CD47 on all other cells. These macrophages were then systemically injected into mice with fluorescent human tumors that had been antibody targeted. Within days, the tumors regressed, and single-cell fluorescence analyses showed that the more the macrophages engulfed, the more they accumulated within regressing tumors. Human-marrow-derived macrophages engorged on the human tumors, while TAMs were minimally phagocytic, even toward CD47-knockdown tumors. Past studies had opsonized tumors in situ with antibody and/or relied on mouse TAMs but had not injected SIRPα-inhibited cells; also, unlike past injections of anti-CD47, blood parameters remained normal and safe. Consistent with tumor-selective engorge-and-accumulate processes in vivo, phagocytosis in vitro inhibited macrophage migration through micropores that mimic features of dense 3D tissue. Accumulation of SIRPα-inhibited macrophages in tumors favored tumor regression for 1-2 weeks, but donor macrophages quickly differentiated toward non-phagocytic, high-SIRPα TAMs. Analyses of macrophages on soft (like marrow) or stiff (like solid tumors) collagenous gels demonstrated a stiffness-driven, retinoic-acid-modulated upregulation of SIRPα and the mechanosensitive nuclear marker lamin-A. Mechanosensitive differentiation was similarly evident in vivo and likely limited the anti-tumor effects, as confirmed by re-initiation of tumor regression by fresh injections of SIRPα-inhibited macrophages. Macrophage motility, phagocytosis, and differentiation in vivo are thus coupled.
Collapse
Affiliation(s)
- Cory M Alvey
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Pharmacological Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyle R Spinler
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerome Irianto
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charlotte R Pfeifer
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon Hayes
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuntao Xia
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sangkyun Cho
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - P C P Dave Dingal
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jake Hsu
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lucas Smith
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Manu Tewari
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Pharmacological Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
308
|
Edwards EE, Oh J, Anilkumar A, Birmingham KG, Thomas SN. P-, but not E- or L-, selectin-mediated rolling adhesion persistence in hemodynamic flow diverges between metastatic and leukocytic cells. Oncotarget 2017; 8:83585-83601. [PMID: 29137366 PMCID: PMC5663538 DOI: 10.18632/oncotarget.18786] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/19/2017] [Indexed: 12/20/2022] Open
Abstract
The ability of leukocytic cells to engage selectins via rolling adhesion is critical to inflammation, but selectins are also implicated in mediating metastatic dissemination. Using a microfluidic- and flow-based cell adhesion chromatography experimental and analytical technique, we interrogated the cell-subtype differences in engagement and sustainment of rolling adhesion on P-, E-, and L-selectin-functionalized surfaces in physiological flow. Our results indicate that, particularly at low concentrations of P-selectin, metastatic but not leukocytic cells exhibit reduced rolling adhesion persistence, whereas both cell subtypes exhibited reduced persistence on L-selectin and high persistence on E-selectin, differences not revealed by flow cytometry analysis or reflected in the extent or velocity of rolling adhesion. Conditions under which adhesion persistence was found to be significantly reduced corresponded to those exhibiting the greatest sensitivity to a selectin-antagonist. Our results suggest that potentially therapeutically exploitable differences in metastatic and leukocytic cell subtype interactions with selectins in physiological flow are identifiable through implementation of functional assays of adhesion persistence in hemodynamic flow utilizing this integrated, flow-based cell adhesion chromatography analytical technique.
Collapse
Affiliation(s)
- Erin Elizabeth Edwards
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Jaeho Oh
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Ananyaveena Anilkumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Katherine Gayle Birmingham
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Susan Napier Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
309
|
Lodygin D, Flügel A. Intravital real-time analysis of T-cell activation in health and disease. Cell Calcium 2017; 64:118-129. [DOI: 10.1016/j.ceca.2016.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 01/27/2023]
|
310
|
Lim JJ, Grinstein S, Roth Z. Diversity and Versatility of Phagocytosis: Roles in Innate Immunity, Tissue Remodeling, and Homeostasis. Front Cell Infect Microbiol 2017; 7:191. [PMID: 28589095 PMCID: PMC5440456 DOI: 10.3389/fcimb.2017.00191] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/03/2017] [Indexed: 12/23/2022] Open
Abstract
Phagocytosis, a critical early event in the microbicidal response of neutrophils, is now appreciated to serve multiple functions in a variety of cell types. Professional phagocytes play a central role in innate immunity by eliminating pathogenic bacteria, fungi and malignant cells, and contribute to adaptive immunity by presenting antigens to lymphocytes. In addition, phagocytes play a part in tissue remodeling and maintain overall homeostasis by disposing of apoptotic cells, a task shared by non-professional phagocytes, often of epithelial origin. This functional versatility is supported by a vast array of receptors capable of recognizing a striking variety of foreign and endogenous ligands. Here we present an abbreviated overview of the different types of phagocytes, their varied modes of signaling and particle engulfment, and the multiple physiological roles of phagocytosis.
Collapse
Affiliation(s)
- Justin J Lim
- Program in Cell Biology, Hospital for Sick ChildrenToronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Hospital for Sick ChildrenToronto, ON, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's HospitalToronto, ON, Canada.,Department of Biochemistry, University of TorontoToronto, ON, Canada
| | - Ziv Roth
- Program in Cell Biology, Hospital for Sick ChildrenToronto, ON, Canada
| |
Collapse
|
311
|
Lavin Y, Kobayashi S, Leader A, Amir EAD, Elefant N, Bigenwald C, Remark R, Sweeney R, Becker CD, Levine JH, Meinhof K, Chow A, Kim-Shulze S, Wolf A, Medaglia C, Li H, Rytlewski JA, Emerson RO, Solovyov A, Greenbaum BD, Sanders C, Vignali M, Beasley MB, Flores R, Gnjatic S, Pe'er D, Rahman A, Amit I, Merad M. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 2017; 169:750-765.e17. [PMID: 28475900 PMCID: PMC5737939 DOI: 10.1016/j.cell.2017.04.014] [Citation(s) in RCA: 895] [Impact Index Per Article: 111.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/26/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022]
Abstract
To guide the design of immunotherapy strategies for patients with early stage lung tumors, we developed a multiscale immune profiling strategy to map the immune landscape of early lung adenocarcinoma lesions to search for tumor-driven immune changes. Utilizing a barcoding method that allows a simultaneous single-cell analysis of the tumor, non-involved lung, and blood cells, we provide a detailed immune cell atlas of early lung tumors. We show that stage I lung adenocarcinoma lesions already harbor significantly altered T cell and NK cell compartments. Moreover, we identified changes in tumor-infiltrating myeloid cell (TIM) subsets that likely compromise anti-tumor T cell immunity. Paired single-cell analyses thus offer valuable knowledge of tumor-driven immune changes, providing a powerful tool for the rational design of immune therapies. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Yonit Lavin
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Soma Kobayashi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew Leader
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - El-Ad David Amir
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Naama Elefant
- Department of Immunology, Weizmann Institute, Rehovot 76100, Israel
| | - Camille Bigenwald
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Romain Remark
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sweeney
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christian D Becker
- Division of Pulmonology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jacob H Levine
- Computational and Systems Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Klaus Meinhof
- Division of Pulmonology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew Chow
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Seunghee Kim-Shulze
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrea Wolf
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chiara Medaglia
- Department of Immunology, Weizmann Institute, Rehovot 76100, Israel
| | - Hanjie Li
- Department of Immunology, Weizmann Institute, Rehovot 76100, Israel
| | | | | | - Alexander Solovyov
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Benjamin D Greenbaum
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | - Mary Beth Beasley
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Raja Flores
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Adeeb Rahman
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ido Amit
- Department of Immunology, Weizmann Institute, Rehovot 76100, Israel
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
312
|
Tatham KC, O'Dea KP, Romano R, Donaldson HE, Wakabayashi K, Patel BV, Thakuria L, Simon AR, Sarathchandra P, Marczin N, Takata M. Intravascular donor monocytes play a central role in lung transplant ischaemia-reperfusion injury. Thorax 2017; 73:350-360. [PMID: 28389600 PMCID: PMC5870457 DOI: 10.1136/thoraxjnl-2016-208977] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 02/28/2017] [Accepted: 03/10/2017] [Indexed: 12/22/2022]
Abstract
Rationale Primary graft dysfunction in lung transplant recipients derives from the initial, largely leukocyte-dependent, ischaemia-reperfusion injury. Intravascular lung-marginated monocytes have been shown to play key roles in experimental acute lung injury, but their contribution to lung ischaemia-reperfusion injury post transplantation is unknown. Objective To define the role of donor intravascular monocytes in lung transplant-related acute lung injury and primary graft dysfunction. Methods Isolated perfused C57BL/6 murine lungs were subjected to warm ischaemia (2 hours) and reperfusion (2 hours) under normoxic conditions. Monocyte retention, activation phenotype and the effects of their depletion by intravenous clodronate-liposome treatment on lung inflammation and injury were determined. In human donor lung transplant samples, the presence and activation phenotype of monocytic cells (low side scatter, 27E10+, CD14+, HLA-DR+, CCR2+) were evaluated by flow cytometry and compared with post-implantation lung function. Results In mouse lungs following ischaemia-reperfusion, substantial numbers of lung-marginated monocytes remained within the pulmonary microvasculature, with reduced L-selectin and increased CD86 expression indicating their activation. Monocyte depletion resulted in reductions in lung wet:dry ratios, bronchoalveolar lavage fluid protein, and perfusate levels of RAGE, MIP-2 and KC, while monocyte repletion resulted in a partial restoration of the injury. In human lungs, correlations were observed between pre-implantation donor monocyte numbers/their CD86 and TREM-1 expression and post-implantation lung dysfunction at 48 and 72 hours. Conclusions These results indicate that lung-marginated intravascular monocytes are retained as a ‘passenger’ leukocyte population during lung transplantation, and play a key role in the development of transplant-associated ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Kate Colette Tatham
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Kieran Patrick O'Dea
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Rosalba Romano
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.,Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Hannah Elizabeth Donaldson
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Kenji Wakabayashi
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Brijesh Vipin Patel
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Louit Thakuria
- Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Andre Rudiger Simon
- Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Padmini Sarathchandra
- Faculty of Medicine, National Heart & Lung Institute, Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middlesex, UK
| | | | - Nandor Marczin
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.,Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Masao Takata
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
313
|
Mechanisms governing metastatic dormancy in breast cancer. Semin Cancer Biol 2017; 44:72-82. [PMID: 28344165 DOI: 10.1016/j.semcancer.2017.03.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023]
Abstract
Breast cancer is a systemic disease characterized by early dissemination of tumor cells to distant organs. In this foreign environment, tumor cells may stay in a dormant state as single cells or as micrometastases for many years before growing out into a macrometastatic lesion. As metastasis is the primary cause for breast cancer-related death, it is important to understand the mechanisms underlying the maintenance of dormancy and dormancy escape to find druggable targets to eradicate metastatic tumor cells. Metastatic dormancy is regulated by complex interactions between tumor cells and the local microenvironment. In addition, cancer-directed immunity and systemic instigation play a crucial role.
Collapse
|
314
|
Quintar A, McArdle S, Wolf D, Marki A, Ehinger E, Vassallo M, Miller J, Mikulski Z, Ley K, Buscher K. Endothelial Protective Monocyte Patrolling in Large Arteries Intensified by Western Diet and Atherosclerosis. Circ Res 2017; 120:1789-1799. [PMID: 28302649 PMCID: PMC5446289 DOI: 10.1161/circresaha.117.310739] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 01/13/2023]
Abstract
Supplemental Digital Content is available in the text. Rationale: Nonclassical mouse monocyte (CX3CR1high, Ly-6Clow) patrolling along the vessels of the microcirculation is critical for endothelial homeostasis and inflammation. Because of technical challenges, it is currently not established how patrolling occurs in large arteries. Objective: This study was undertaken to elucidate the molecular, migratory, and functional phenotypes of patrolling monocytes in the high shear and pulsatile environment of large arteries in healthy, hyperlipidemic, and atherosclerotic conditions. Methods and Results: Applying a new method for stable, long-term 2-photon intravital microscopy of unrestrained large arteries in live CX3CR1-GFP (green fluorescent protein) mice, we show that nonclassical monocytes patrol inside healthy carotid arteries at a velocity of 36 μm/min, 3× faster than in microvessels. The tracks are less straight but lead preferentially downstream. The number of patrolling monocytes is increased 9-fold by feeding wild-type mice a Western diet or by applying topical TLR7/8 (Toll-like receptor) agonists. A similar increase is seen in CX3CR1+/GFP/apoE−/− mice on chow diet, with a further 2- to 3-fold increase on Western diet (22-fold over healthy). In plaque conditions, monocytes are readily captured onto the endothelium from free flow. Stable patrolling is unaffected in CX3CR1-deficient mice and involves the contribution of LFA-1 (lymphocyte-associated antigen 1) and α4 integrins. The endothelial damage in atherosclerotic carotid arteries was assessed by electron microscopy and correlates with the number of intraluminal patrollers. Abolishing patrolling monocytes in Nr4a1−/− apoE−/− mice leads to pronounced endothelial apoptosis. Conclusions: Arterial patrolling is a prominent new feature of nonclassical monocytes with unique molecular and kinetic properties. It is highly upregulated in hyperlipidemia and atherosclerosis in a CX3CR1-independent fashion and plays a potential role in endothelial protection.
Collapse
Affiliation(s)
- Amado Quintar
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Sara McArdle
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Dennis Wolf
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Alex Marki
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Erik Ehinger
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Melanie Vassallo
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Jacqueline Miller
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Zbigniew Mikulski
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Klaus Ley
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.)
| | - Konrad Buscher
- From the La Jolla Institute for Allergy and Immunology, Division of Inflammation Biology, La Jolla, CA (A.Q., S.M., D.W., A.M., E.E., M.V., J.M., Z.M., K.L., K.B.); and Centro de Microscopia Electronica, INICSA-CONICET, Facultad de Ciencias Medicas, Universidad Nacional de Cordoba, Cordoba, Argentina (A.Q.).
| |
Collapse
|
315
|
Rahman MS, Murphy AJ, Woollard KJ. Effects of dyslipidaemia on monocyte production and function in cardiovascular disease. Nat Rev Cardiol 2017; 14:387-400. [PMID: 28300081 DOI: 10.1038/nrcardio.2017.34] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Monocytes are heterogeneous effector cells involved in the maintenance and restoration of tissue integrity. Monocytes and macrophages are involved in cardiovascular disease progression, and are associated with the development of unstable atherosclerotic plaques. Hyperlipidaemia can accelerate cardiovascular disease progression. However, monocyte responses to hyperlipidaemia are poorly understood. In the past decade, accumulating data describe the relationship between the dynamic blood lipid environment and the heterogeneous circulating monocyte pool, which might have profound consequences for cardiovascular disease. In this Review, we explore the updated view of monocytes in cardiovascular disease and their relationship with macrophages in promoting the homeostatic and inflammatory responses related to atherosclerosis. We describe the different definitions of dyslipidaemia, highlight current theories on the ontogeny of monocyte heterogeneity, discuss how dyslipidaemia might alter monocyte production, and explore the mechanistic interface linking dyslipidaemia with monocyte effector functions, such as migration and the inflammatory response. Finally, we discuss the role of dietary and endogenous lipid species in mediating dyslipidaemic responses, and the role of these lipids in promoting the risk of cardiovascular disease through modulation of monocyte behaviour.
Collapse
Affiliation(s)
- Mohammed Shamim Rahman
- Renal &Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology Lab, Baker IDI Heart &Diabetes Research Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Department of Immunology, Monash University, 89 Commercial Road, Melbourne, Victoria 3004, Australia
| | - Kevin J Woollard
- Renal &Vascular Inflammation Section, Division of Immunology and Inflammation, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
316
|
Kelleher FC, O'Sullivan H. Monocytes, Macrophages, and Osteoclasts in Osteosarcoma. J Adolesc Young Adult Oncol 2017; 6:396-405. [PMID: 28263668 DOI: 10.1089/jayao.2016.0078] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Macrophages appear to have a fundamental role in the pathogenesis of osteosarcoma. These highly diverse plastic cells are subdivided into classical or inflammatory macrophages known as M1 and alternative macrophages, which decrease inflammation and are reparative, called M2. Although primary and metastatic osteosarcomas are infiltrated with M2 macrophages, targeting the M1 macrophages with the immune adjuvant muramyl tripeptide phosphatidyl ethanolamine (MTP-PE) has been the greatest recent therapeutic advance in osteosarcoma. This discrepancy between the presence of M2 and activation of M1 macrophages is intriguing and is likely explained either by the plasticity of M1 and M2 macrophages or nonclassical patrolling monocytes (PMos). To date, MTP-PE has been approved in combination with chemotherapy for nonmetastatic osteosarcoma, but its use in metastatic tumors has not been investigated. In this review, we focus on macrophages, monocytes, and osteoclasts, their role in osteosarcoma, and the potential for targeting these cells in this disease.
Collapse
Affiliation(s)
- Fergal C Kelleher
- 1 Trinity College Dublin , Dublin, Ireland .,2 Department of Medical Oncology, St. James Hospital , Dublin, Ireland
| | - Hazel O'Sullivan
- 2 Department of Medical Oncology, St. James Hospital , Dublin, Ireland .,3 Whangarei Base Hospital , Whangarei, New Zealand
| |
Collapse
|
317
|
Li XM, Wang JR, Shen T, Gao SS, He XS, Li JN, Yang TY, Zhang S, Gan WJ, Li JM, Wu H. Nur77 deficiency in mice accelerates tumor invasion and metastasis by facilitating TNFα secretion and lowering CSF-1R expression. PLoS One 2017; 12:e0171347. [PMID: 28170411 PMCID: PMC5295676 DOI: 10.1371/journal.pone.0171347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/19/2017] [Indexed: 12/26/2022] Open
Abstract
Nur77, an orphan member of the nuclear receptor superfamily, plays critical roles in inflammation and immunity. However, the role of Nur77 in tumor microenvironment remains elusive. Results showed that deletion of Nur77 strikingly enhanced tumor metastasis compared to WT mice. Additionally, compared to the conditioned media derived from Nur77+/+ peritoneal macrophages (CM1), the conditioned media derived from Nur77-/- peritoneal macrophages (CM2) significantly promoted the EMT of cancer cells, and greatly enhanced the migratory and invasive abilities of cancer cells. Moreover, studies using TNF-α blocking antibody demonstrated that pro-inflammatory cytokine TNF-α was indispensable in supporting CM2-induced EMT to drive cancer cells migration and invasion. Furthermore, we found that Nur77 promoted the expression of CSF-1R, a novel downstream target gene of Nur77, and subsequently enhanced the migration of inflammatory cells. Notably, infiltration of inflammatory cells in the tumors of Nur77-/- mice was markedly abrogated compared to Nur77+/+ mice. Collectively, these results revealed that host Nur77 expression was pivotal in antitumor immune response, and in inhibiting tumor metastasis.
Collapse
Affiliation(s)
- Xiu-Ming Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Jing-Ru Wang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Tong Shen
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Shang-Shang Gao
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Xiao-Shun He
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiang-Nan Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Tian-Yu Yang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Shen Zhang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Wen-Juan Gan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- * E-mail: (HW); (JML)
| | - Hua Wu
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
- * E-mail: (HW); (JML)
| |
Collapse
|
318
|
78495111110.1038/nrclinonc.2016.217" />
|
319
|
Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 2017; 14:399-416. [PMID: 28117416 DOI: 10.1038/nrclinonc.2016.217] [Citation(s) in RCA: 2781] [Impact Index Per Article: 347.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Macrophages are crucial drivers of tumour-promoting inflammation. Tumour-associated macrophages (TAMs) contribute to tumour progression at different levels: by promoting genetic instability, nurturing cancer stem cells, supporting metastasis, and taming protective adaptive immunity. TAMs can exert a dual, yin-yang influence on the effectiveness of cytoreductive therapies (chemotherapy and radiotherapy), either antagonizing the antitumour activity of these treatments by orchestrating a tumour-promoting, tissue-repair response or, instead, enhancing the overall antineoplastic effect. TAMs express molecular triggers of checkpoint proteins that regulate T-cell activation, and are targets of certain checkpoint-blockade immunotherapies. Other macrophage-centred approaches to anticancer therapy are under investigation, and include: inhibition of macrophage recruitment to, and/or survival in, tumours; functional re-education of TAMs to an antitumour, 'M1-like' mode; and tumour-targeting monoclonal antibodies that elicit macrophage-mediated extracellular killing, or phagocytosis and intracellular destruction of cancer cells. The evidence supporting these strategies is reviewed herein. We surmise that TAMs can provide tools to tailor the use of cytoreductive therapies and immunotherapy in a personalized medicine approach, and that TAM-focused therapeutic strategies have the potential to complement and synergize with both chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Alberto Mantovani
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy.,Humanitas University, Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Federica Marchesi
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20133 Milan, Italy
| | - Alberto Malesci
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20133 Milan, Italy
| | - Luigi Laghi
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| | - Paola Allavena
- Istituto Clinico Humanitas, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Via A. Manzoni 113, 20089 Rozzano, Milan, Italy.,Humanitas University, Via A. Manzoni 113, 20089 Rozzano, Milan, Italy
| |
Collapse
|
320
|
Mononuclear phagocytes as a target, not a barrier, for drug delivery. J Control Release 2017; 259:53-61. [PMID: 28108325 DOI: 10.1016/j.jconrel.2017.01.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/06/2017] [Accepted: 01/16/2017] [Indexed: 02/07/2023]
Abstract
Mononuclear phagocytes have been generally recognized as a barrier to drug delivery. Recently, a new understanding of mononuclear phagocytes (MPS) ontogeny has surfaced and their functions in disease have been unveiled, demonstrating the need for re-evaluation of perspectives on mononuclear phagocytes in drug delivery. In this review, we described mononuclear phagocyte biology and focus on their accumulation mechanisms in disease sites with explanations of monocyte heterogeneity. In the 'MPS as a barrier' section, we summarized recent studies on mechanisms to avoid phagocytosis based on two different biological principles: protein adsorption and self-recognition. In the 'MPS as a target' section, more detailed descriptions were given on mononuclear phagocyte-targeted drug delivery systems and their applications to various diseases. Collectively, we emphasize in this review that mononuclear phagocytes are potent targets for future drug delivery systems. Mononuclear phagocyte-targeted delivery systems should be created with an understanding of mononuclear phagocyte ontogeny and pathology. Each specific subset of phagocytes should be targeted differently by location and function for improved disease-drug delivery while avoiding RES clearance such as Kupffer cells and splenic macrophages.
Collapse
|
321
|
Lehmann B, Biburger M, Brückner C, Ipsen-Escobedo A, Gordan S, Lehmann C, Voehringer D, Winkler T, Schaft N, Dudziak D, Sirbu H, Weber GF, Nimmerjahn F. Tumor location determines tissue-specific recruitment of tumor-associated macrophages and antibody-dependent immunotherapy response. Sci Immunol 2017; 2:2/7/eaah6413. [PMID: 28783667 DOI: 10.1126/sciimmunol.aah6413] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/08/2016] [Indexed: 12/14/2022]
Abstract
Despite recent advances in activating immune cells to target tumors, the presence of some immune cells, such as tumor-associated macrophages (TAMs) or tumor-associated neutrophils (TANs), may promote rather than inhibit tumor growth. However, it remains unclear how antibody-dependent tumor immunotherapies, such as cytotoxic or checkpoint control antibodies, affect different TAM or TAN populations, which abundantly express activating Fcγ receptors. In this study, we show that the tissue environment determines which cellular effector pathways are responsible for antibody-dependent tumor immunotherapy. Although TAMs derived from Ly6Chigh monocytes recruited by the CCL2-CCR2 axis were critical for tumor immunotherapy of skin tumors, the destruction of lung tumors was CCL2-independent and required the presence of colony-stimulating factor 2-dependent tissue-resident macrophages. Our findings suggest that TAMs may have a dual role not only in promoting tumor growth in certain tissue environments on the one hand but also in contributing to tumor cell destruction during antibody-mediated immunotherapy on the other hand.
Collapse
Affiliation(s)
- Birgit Lehmann
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany
| | - Markus Biburger
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany
| | - Christin Brückner
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany
| | - Andrea Ipsen-Escobedo
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany
| | - Sina Gordan
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany
| | - Christian Lehmann
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen, Wasserturmstr. 3/5, 91054 Erlangen, Germany
| | - Thomas Winkler
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Horia Sirbu
- Department of Surgery, University Hospital Erlangen, Krankenhausstr. 12, 91054 Erlangen, Germany
| | - Georg F Weber
- Department of Surgery, University Hospital Erlangen, Krankenhausstr. 12, 91054 Erlangen, Germany
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erwin-Rommel-Str. 3, 91058 Erlangen, Germany.
| |
Collapse
|
322
|
Tumor Associated Macrophages as Therapeutic Targets for Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:331-370. [PMID: 29282692 DOI: 10.1007/978-981-10-6020-5_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor-associated macrophages (TAMs) are the most abundant inflammatory infiltrates in the tumor stroma. TAMs promote tumor growth by suppressing immunocompetent cells, including neovascularization and supporting cancer stem cells. In the chapter, we discuss recent efforts in reprogramming or inhibiting tumor-protecting properties of TAMs, and developing potential strategies to increase the efficacy of breast cancer treatment.
Collapse
|
323
|
Novel insights in the regulation and function of macrophages in the tumor microenvironment. Curr Opin Oncol 2017; 29:55-61. [DOI: 10.1097/cco.0000000000000344] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
324
|
Day TF, Mewani RR, Starr J, Li X, Chakravarty D, Ressom H, Zou X, Eidelman O, Pollard HB, Srivastava M, Kasid UN. Transcriptome and Proteome Analyses of TNFAIP8 Knockdown Cancer Cells Reveal New Insights into Molecular Determinants of Cell Survival and Tumor Progression. Methods Mol Biol 2017; 1513:83-100. [PMID: 27807832 DOI: 10.1007/978-1-4939-6539-7_7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Tumor necrosis factor-α-inducible protein 8 (TNFAIP8) is the first discovered oncogenic and an anti-apoptotic member of a conserved TNFAIP8 or TIPE family of proteins. TNFAIP8 mRNA is induced by NF-kB, and overexpression of TNFAIP8 has been correlated with poor prognosis in many cancers. Downregulation of TNFAIP8 expression has been associated with decreased pulmonary colonization of human tumor cells, and enhanced sensitivities of tumor xenografts to radiation and docetaxel. Here we have investigated the effects of depletion of TNFAIP8 on the mRNA, microRNA and protein expression profiles in prostate and breast cancers and melanoma. Depending on the tumor cell type, knockdown of TNFAIP8 was found to be associated with increased mRNA expression of several antiproliferative and apoptotic genes (e.g., IL-24, FAT3, LPHN2, EPHA3) and fatty acid oxidation gene ACADL, and decreased mRNA levels of oncogenes (e.g., NFAT5, MALAT1, MET, FOXA1, KRAS, S100P, OSTF1) and glutamate transporter gene SLC1A1. TNFAIP8 knockdown cells also exhibited decreased expression of multiple onco-proteins (e.g., PIK3CA, SRC, EGFR, IL5, ABL1, GAP43), and increased expression of the orphan nuclear receptor NR4A1 and alpha 1 adaptin subunit of the adaptor-related protein complex 2 AP2 critical to clathrin-mediated endocytosis. TNFAIP8-centric molecules were found to be predominately implicated in the hypoxia-inducible factor-1α (HIF-1α) signaling pathway, and cancer and development signaling networks. Thus TNFAIP8 seems to regulate the cell survival and cancer progression processes in a multifaceted manner. Future validation of the molecules identified in this study is likely to lead to new subset of molecules and functional determinants of cancer cell survival and progression.
Collapse
Affiliation(s)
- Timothy F Day
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Rajshree R Mewani
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Joshua Starr
- Department of Anatomy, Physiology and Genetics, Institute for Molecular Medicine, Center for Medical Proteomics, Uniformed Services University School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Xin Li
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Debyani Chakravarty
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Habtom Ressom
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Xiaojun Zou
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Ofer Eidelman
- Department of Anatomy, Physiology and Genetics, Institute for Molecular Medicine, Center for Medical Proteomics, Uniformed Services University School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Harvey B Pollard
- Department of Anatomy, Physiology and Genetics, Institute for Molecular Medicine, Center for Medical Proteomics, Uniformed Services University School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, Institute for Molecular Medicine, Center for Medical Proteomics, Uniformed Services University School of Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Usha N Kasid
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
325
|
Liu Y, Cao X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell 2016; 30:668-681. [PMID: 27846389 DOI: 10.1016/j.ccell.2016.09.011] [Citation(s) in RCA: 806] [Impact Index Per Article: 89.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/22/2016] [Accepted: 09/20/2016] [Indexed: 12/17/2022]
Abstract
Primary tumors create a favorable microenvironment, namely, pre-metastatic niche, in secondary organs and tissue sites for subsequent metastases. The pre-metastatic niche can be primed and established through a complex interplay among primary tumor-derived factors, tumor-mobilized bone marrow-derived cells, and local stromal components. We review here our current understanding of the key components and underlying mechanisms for pre-metastatic niche formation. We propose six characteristics that define the pre-metastatic niche, which enable tumor cell colonization and promote metastasis, including immunosuppression, inflammation, angiogenesis/vascular permeability, lymphangiogenesis, organotropism, and reprogramming. We highlight the significance of the pre-metastatic niche, and discuss potential implications and future research directions.
Collapse
Affiliation(s)
- Yang Liu
- National Key Laboratory of Medical Molecular Biology, Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xuetao Cao
- National Key Laboratory of Medical Molecular Biology, Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
326
|
Heterogeneity, functional specialization and differentiation of monocyte‐derived dendritic cells. Immunol Cell Biol 2016; 95:244-251. [DOI: 10.1038/icb.2016.104] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 02/07/2023]
|
327
|
Li X, Fang P, Yang WY, Chan K, Lavallee M, Xu K, Gao T, Wang H, Yang X. Mitochondrial ROS, uncoupled from ATP synthesis, determine endothelial activation for both physiological recruitment of patrolling cells and pathological recruitment of inflammatory cells. Can J Physiol Pharmacol 2016; 95:247-252. [PMID: 27925481 DOI: 10.1139/cjpp-2016-0515] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mitochondrial reactive oxygen species (mtROS) are signaling molecules, which drive inflammatory cytokine production and T cell activation. In addition, cardiovascular diseases, cancers, and autoimmune diseases all share a common feature of increased mtROS level. Both mtROS and ATP are produced as a result of electron transport chain activity, but it remains enigmatic whether mtROS could be generated independently from ATP synthesis. A recent study shed light on this important question and found that, during endothelial cell (EC) activation, mtROS could be upregulated in a proton leak-coupled, but ATP synthesis-uncoupled manner. As a result, EC could upregulate mtROS production for physiological EC activation without compromising mitochondrial membrane potential and ATP generation, and consequently without causing mitochondrial damage and EC death. Thus, a novel pathophysiological role of proton leak in driving mtROS production was uncovered for low grade EC activation, patrolling immunosurveillance cell trans-endothelial migration and other signaling events without compromising cellular survival. This new working model explains how mtROS could be increasingly generated independently from ATP synthesis and endothelial damage or death. Mapping the connections among mitochondrial metabolism, physiological EC activation, patrolling cell migration, and pathological inflammation is significant towards the development of novel therapies for inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Xinyuan Li
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Pu Fang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - William Y Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Kylie Chan
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Muriel Lavallee
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Keman Xu
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Tracy Gao
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
328
|
Thomas GD, Hanna RN, Vasudevan NT, Hamers AA, Romanoski CE, McArdle S, Ross KD, Blatchley A, Yoakum D, Hamilton BA, Mikulski Z, Jain MK, Glass CK, Hedrick CC. Deleting an Nr4a1 Super-Enhancer Subdomain Ablates Ly6C low Monocytes while Preserving Macrophage Gene Function. Immunity 2016; 45:975-987. [PMID: 27814941 DOI: 10.1016/j.immuni.2016.10.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/12/2016] [Accepted: 08/15/2016] [Indexed: 01/08/2023]
Abstract
Mononuclear phagocytes are a heterogeneous family that occupy all tissues and assume numerous roles to support tissue function and systemic homeostasis. Our ability to dissect the roles of individual subsets is limited by a lack of technologies that ablate gene function within specific mononuclear phagocyte sub-populations. Using Nr4a1-dependent Ly6Clow monocytes, we present a proof-of-principle approach that addresses these limitations. Combining ChIP-seq and molecular approaches we identified a single, conserved, sub-domain within the Nr4a1 enhancer that was essential for Ly6Clow monocyte development. Mice lacking this enhancer lacked Ly6Clow monocytes but retained Nr4a1 gene expression in macrophages during steady state and in response to LPS. Because Nr4a1 regulates inflammatory gene expression and differentiation of Ly6Clow monocytes, decoupling these processes allows Ly6Clow monocytes to be studied independently.
Collapse
Affiliation(s)
- Graham D Thomas
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Richard N Hanna
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Neelakatan T Vasudevan
- Case Cardiovascular Research Institute, Department of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Anouk A Hamers
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Casey E Romanoski
- Keating Bioresearch Building, 1657 E. Helen St, Tucson, AZ 85721, USA
| | - Sara McArdle
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Kevin D Ross
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Amy Blatchley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Deborah Yoakum
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Bruce A Hamilton
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Zbigniew Mikulski
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Department of Medicine, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
329
|
Hanna RN, Hedrick CC. Quantification of Tumor Material Uptake. Bio Protoc 2016; 6:e1974. [PMID: 29552586 DOI: 10.21769/bioprotoc.1974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Extracellular tumor material including exosomes, microvesicles and apoptotic tumor debris may help cancers invade new organs. Enhancing the removal of extracellular tumor material by immune cells represents a novel immunotherapy approach for preventing cancer metastasis. This protocol quantifies the uptake and removal of extracellular tumor material from circulation and tissues by immune cells. In this assay fluorescent tumor cells are transferred into mice, and then immune cells are quantified by either flow cytometry or imaging cytometry for their uptake of tumor material.
Collapse
Affiliation(s)
- Richard N Hanna
- Department of Respiratory, Inflammation and Autoimmune Diseases, MedImmune, LLC, Gaithersburg, USA.,Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, USA
| |
Collapse
|
330
|
Kratofil RM, Kubes P, Deniset JF. Monocyte Conversion During Inflammation and Injury. Arterioscler Thromb Vasc Biol 2016; 37:35-42. [PMID: 27765768 DOI: 10.1161/atvbaha.116.308198] [Citation(s) in RCA: 322] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/10/2016] [Indexed: 12/25/2022]
Abstract
Monocytes are circulating leukocytes important in both innate and adaptive immunity, primarily functioning in immune defense, inflammation, and tissue remodeling. There are 2 subsets of monocytes in mice (3 subsets in humans) that are mobilized from the bone marrow and recruited to sites of inflammation, where they carry out their respective functions in promoting inflammation or facilitating tissue repair. Our understanding of the fate of these monocyte subsets at the site of inflammation is constantly evolving. This brief review highlights the plasticity of monocyte subsets and their conversion during inflammation and injury.
Collapse
Affiliation(s)
- Rachel M Kratofil
- From the Department of Microbiology, Immunology, and Infectious Diseases (R.M.K., P.K.) and Department of Physiology and Pharmacology (P.K., J.F.D.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Canada
| | - Paul Kubes
- From the Department of Microbiology, Immunology, and Infectious Diseases (R.M.K., P.K.) and Department of Physiology and Pharmacology (P.K., J.F.D.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Canada
| | - Justin F Deniset
- From the Department of Microbiology, Immunology, and Infectious Diseases (R.M.K., P.K.) and Department of Physiology and Pharmacology (P.K., J.F.D.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Canada
| |
Collapse
|
331
|
Chong SZ, Evrard M, Devi S, Chen J, Lim JY, See P, Zhang Y, Adrover JM, Lee B, Tan L, Li JLY, Liong KH, Phua C, Balachander A, Boey A, Liebl D, Tan SM, Chan JKY, Balabanian K, Harris JE, Bianchini M, Weber C, Duchene J, Lum J, Poidinger M, Chen Q, Rénia L, Wang CI, Larbi A, Randolph GJ, Weninger W, Looney MR, Krummel MF, Biswas SK, Ginhoux F, Hidalgo A, Bachelerie F, Ng LG. CXCR4 identifies transitional bone marrow premonocytes that replenish the mature monocyte pool for peripheral responses. J Exp Med 2016; 213:2293-2314. [PMID: 27811056 PMCID: PMC5068243 DOI: 10.1084/jem.20160800] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/01/2016] [Indexed: 11/04/2022] Open
Abstract
It is well established that Ly6Chi monocytes develop from common monocyte progenitors (cMoPs) and reside in the bone marrow (BM) until they are mobilized into the circulation. In our study, we found that BM Ly6Chi monocytes are not a homogenous population, as current data would suggest. Using computational analysis approaches to interpret multidimensional datasets, we demonstrate that BM Ly6Chi monocytes consist of two distinct subpopulations (CXCR4hi and CXCR4lo subpopulations) in both mice and humans. Transcriptome studies and in vivo assays revealed functional differences between the two subpopulations. Notably, the CXCR4hi subset proliferates and is immobilized in the BM for the replenishment of functionally mature CXCR4lo monocytes. We propose that the CXCR4hi subset represents a transitional premonocyte population, and that this sequential step of maturation from cMoPs serves to maintain a stable pool of BM monocytes. Additionally, reduced CXCR4 expression on monocytes, upon their exit into the circulation, does not reflect its diminished role in monocyte biology. Specifically, CXCR4 regulates monocyte peripheral cellular activities by governing their circadian oscillations and pulmonary margination, which contributes toward lung injury and sepsis mortality. Together, our study demonstrates the multifaceted role of CXCR4 in defining BM monocyte heterogeneity and in regulating their function in peripheral tissues.
Collapse
Affiliation(s)
- Shu Zhen Chong
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Maximilien Evrard
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore.,School of Biological Sciences, Nanyang Technological University, 637551 Singapore
| | - Sapna Devi
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Jyue Yuan Lim
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Peter See
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Yiru Zhang
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Biopolis, 138673 Singapore
| | - José M Adrover
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Bernett Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Leonard Tan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Jackson L Y Li
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Ka Hang Liong
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Cindy Phua
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Akhila Balachander
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Adrian Boey
- Institute of Medical Biology (IMB)-Institute of Molecular and Cell Biology (IMCB) Electron Microscopy Suite, A*STAR (Agency for Science, Technology and Research), Biopolis, 138671 Singapore
| | - David Liebl
- Institute of Medical Biology (IMB)-Institute of Molecular and Cell Biology (IMCB) Electron Microscopy Suite, A*STAR (Agency for Science, Technology and Research), Biopolis, 138671 Singapore
| | - Suet Mien Tan
- School of Biological Sciences, Nanyang Technological University, 637551 Singapore
| | - Jerry K Y Chan
- Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore.,Department of Reproductive Medicine, KK Women's and Children's Hospital, 229899 Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, 169857 Singapore
| | - Karl Balabanian
- INSERM UMR-S996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, 92140 Clamart, France
| | - John E Harris
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Mariaelvy Bianchini
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich 80336, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich 80336, Germany
| | - Johan Duchene
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich 80336, Germany
| | - Josephine Lum
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Biopolis, 138673 Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Cheng-I Wang
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | | | - Wolfgang Weninger
- Centenary Institute for Cancer Medicine and Cell Biology, Newton, New South Wales 2042, Australia
| | - Mark R Looney
- Department of Medicine and Pathology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Matthew F Krummel
- Department of Medicine and Pathology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich 80336, Germany
| | - Françoise Bachelerie
- INSERM UMR-S996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, 92140 Clamart, France
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore .,School of Biological Sciences, Nanyang Technological University, 637551 Singapore
| |
Collapse
|
332
|
Fernández-Figueras MT, Martín-Urdà MT, Plana A, Servitje O, Penin RM, Tapia G, Mate JL, Ariza A. Intravascular (blood vessel) histiocytosis with haemophagocytosis. Histopathology 2016; 69:1077-1081. [DOI: 10.1111/his.13030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 07/04/2016] [Indexed: 01/02/2023]
Affiliation(s)
- M Teresa Fernández-Figueras
- Department of Pathology; Hospital Universitari Germans Trias i Pujol; Universitat Autònoma de Barcelona; Badalona Barcelona Spain
| | - M Teresa Martín-Urdà
- Department of Dermatology; Hospital Municipal de Badalona; Badalona Barcelona Spain
| | - Adrià Plana
- Department of Dermatology; Hospital Universitari Germans Trias i Pujol; Universitat Autònoma de Barcelona; Badalona Barcelona Spain
| | - Octavi Servitje
- Department of Dermatology; Hospital Universitari de Bellvitge; Universitat Autònoma de Barcelona; L'Hospitalet de Llobregat; Barcelona Spain
| | - Rosa M Penin
- Department of Pathology; Hospital Universitari de Bellvitge; Universitat Autònoma de Barcelona; L'Hospitalet de Llobregat; Barcelona Spain
| | - Gustavo Tapia
- Department of Pathology; Hospital Universitari Germans Trias i Pujol; Universitat Autònoma de Barcelona; Badalona Barcelona Spain
| | - José L Mate
- Department of Pathology; Hospital Universitari Germans Trias i Pujol; Universitat Autònoma de Barcelona; Badalona Barcelona Spain
| | - Aurelio Ariza
- Department of Pathology; Hospital Universitari Germans Trias i Pujol; Universitat Autònoma de Barcelona; Badalona Barcelona Spain
| |
Collapse
|
333
|
He H, Mack JJ, Güç E, Warren CM, Squadrito ML, Kilarski WW, Baer C, Freshman RD, McDonald AI, Ziyad S, Swartz MA, De Palma M, Iruela-Arispe ML. Perivascular Macrophages Limit Permeability. Arterioscler Thromb Vasc Biol 2016; 36:2203-2212. [PMID: 27634833 DOI: 10.1161/atvbaha.116.307592] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/31/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Perivascular cells, including pericytes, macrophages, smooth muscle cells, and other specialized cell types, like podocytes, participate in various aspects of vascular function. However, aside from the well-established roles of smooth muscle cells and pericytes, the contributions of other vascular-associated cells are poorly understood. Our goal was to ascertain the function of perivascular macrophages in adult tissues under nonpathological conditions. APPROACH AND RESULTS We combined confocal microscopy, in vivo cell depletion, and in vitro assays to investigate the contribution of perivascular macrophages to vascular function. We found that resident perivascular macrophages are associated with capillaries at a frequency similar to that of pericytes. Macrophage depletion using either clodronate liposomes or antibodies unexpectedly resulted in hyperpermeability. This effect could be rescued when M2-like macrophages, but not M1-like macrophages or dendritic cells, were reconstituted in vivo, suggesting subtype-specific roles for macrophages in the regulation of vascular permeability. Furthermore, we found that permeability-promoting agents elicit motility and eventual dissociation of macrophages from the vasculature. Finally, in vitro assays showed that M2-like macrophages attenuate the phosphorylation of VE-cadherin upon exposure to permeability-promoting agents. CONCLUSIONS This study points to a direct contribution of macrophages to vessel barrier integrity and provides evidence that heterotypic cell interactions with the endothelium, in addition to those of pericytes, control vascular permeability.
Collapse
Affiliation(s)
- Huanhuan He
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Julia J Mack
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Esra Güç
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Carmen M Warren
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Mario Leonardo Squadrito
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Witold W Kilarski
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Caroline Baer
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Ryan D Freshman
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Austin I McDonald
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Safiyyah Ziyad
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Melody A Swartz
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Michele De Palma
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - M Luisa Iruela-Arispe
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.).
| |
Collapse
|
334
|
Xin Y, Gao X, Wang W, Xu X, Yu L, Ju X, Li A. Circulating cell-free DNA indicates M1/M2 responses during septic peritonitis. Biochem Biophys Res Commun 2016; 477:589-594. [DOI: 10.1016/j.bbrc.2016.06.092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/10/2016] [Accepted: 06/18/2016] [Indexed: 12/23/2022]
|
335
|
Zhu YP, Thomas GD, Hedrick CC. 2014 Jeffrey M. Hoeg Award Lecture: Transcriptional Control of Monocyte Development. Arterioscler Thromb Vasc Biol 2016; 36:1722-33. [PMID: 27386937 PMCID: PMC5828163 DOI: 10.1161/atvbaha.116.304054] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/24/2016] [Indexed: 01/01/2023]
Abstract
Monocytes and macrophages are key immune cells involved in the early progression of atherosclerosis. Transcription factors that control their development in the bone marrow are important therapeutic targets to control the numbers and functions of these cells in disease. This review highlights what is currently known about the transcription factors that are critical for monocyte development.
Collapse
Affiliation(s)
- Yanfang Peipei Zhu
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Graham D Thomas
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
336
|
Fiole D, Tournier JN. Intravital microscopy of the lung: minimizing invasiveness. JOURNAL OF BIOPHOTONICS 2016; 9:868-878. [PMID: 26846880 DOI: 10.1002/jbio.201500246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 06/05/2023]
Abstract
In vivo microscopy has recently become a gold standard in lung immunology studies involving small animals, largely benefiting from the democratization of multiphoton microscopy allowing for deep tissue imaging. This technology represents currently our only way of exploring the lungs and inferring what happens in human respiratory medicine. The interest of lung in vivo microscopy essentially relies upon its relevance as a study model, fulfilling physiological requirements in comparison with in vitro and ex vivo experiments. However, strategies developed in order to overcome movements of the thorax caused by breathing and heartbeats remain the chief drawback of the technique and a major source of invasiveness. In this context, minimizing invasiveness is an unavoidable prerequisite for any improvement of lung in vivo microscopy. This review puts into perspective the main techniques enabling lung in vivo microscopy, providing pros and cons regarding invasiveness.
Collapse
Affiliation(s)
- Daniel Fiole
- Unité Interactions Hôte-Agents pathogènes, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge cedex, 91223, France.
- Human Histopathology and Animal Models, Institut Pasteur, 28 rue du docteur Roux, Paris, 75725, France.
| | - Jean-Nicolas Tournier
- Unité Interactions Hôte-Agents pathogènes, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge cedex, 91223, France
- Laboratoire Pathogénie des Toxi-Infections Bactériennes, Institut Pasteur, 28 rue du docteur Roux, Paris, 75725, France
- Ecole du Val-de-Grâce, 1 place Alphonse Laveran, Paris, 75230, France
| |
Collapse
|
337
|
Abstract
Cancer heterogeneity, a hallmark enabling clonal survival and therapy resistance, is shaped by active immune responses. Antigen-specific T cells can control cancer, as revealed clinically by immunotherapeutics such as adoptive T-cell transfer and checkpoint blockade. The host immune system is thus a powerful tool that, if better harnessed, could significantly enhance the efficacy of cytotoxic therapy and improve outcomes for cancer sufferers. To realize this vision, however, a number of research frontiers must be tackled. These include developing strategies for neutralizing tumor-promoting inflammation, broadening T-cell repertoires (via vaccination), and elucidating the mechanisms by which immune cells organize tumor microenvironments to regulate T-cell activity. Such efforts will pave the way for identifying new targets for combination therapies that overcome resistance to current treatments and promote long-term cancer control.
Collapse
|
338
|
Affiliation(s)
- Jeffrey W Pollard
- From the MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh
| |
Collapse
|
339
|
Lewis CE, Harney AS, Pollard JW. The Multifaceted Role of Perivascular Macrophages in Tumors. Cancer Cell 2016; 30:18-25. [PMID: 27411586 PMCID: PMC5024543 DOI: 10.1016/j.ccell.2016.05.017] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/18/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Evidence has emerged for macrophages in the perivascular niche of tumors regulating important processes like angiogenesis, various steps in the metastatic cascade, the recruitment and activity of other tumor-promoting leukocytes, and tumor responses to frontline therapies like irradiation and chemotherapy. Understanding the mechanisms controlling the recruitment, retention, and function of these cells could identify important targets for anti-cancer therapeutics.
Collapse
Affiliation(s)
- Claire E Lewis
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield S10 2RX, UK.
| | - Allison S Harney
- Departments of Anatomy and Structural Biology, Developmental and Molecular Biology, Albert Einstein College of Medicine, New York 10461, USA
| | - Jeffrey W Pollard
- Departments of Anatomy and Structural Biology, Developmental and Molecular Biology, Albert Einstein College of Medicine, New York 10461, USA; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
340
|
Abstract
Recent clinical trials have demonstrated the ability to durably control cancer in some patients by manipulating T lymphocytes. These immunotherapies are revolutionizing cancer treatment but benefit only a minority of patients. It is thus a crucial time for clinicians, cancer scientists and immunologists to determine the next steps in shifting cancer treatment towards better cancer control. This Review describes recent advances in our understanding of tumour-associated myeloid cells. These cells remain less studied than T lymphocytes but have attracted particular attention because their presence in tumours is often linked to altered patient survival. Also, experimental studies indicate that myeloid cells modulate key cancer-associated activities, including immune evasion, and affect virtually all types of cancer therapy. Consequently, targeting myeloid cells could overcome limitations of current treatment options.
Collapse
Affiliation(s)
- Camilla Engblom
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
- Graduate Program in Immunology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
341
|
de Mingo Pulido A, Ruffell B. Immune Regulation of the Metastatic Process: Implications for Therapy. Adv Cancer Res 2016; 132:139-63. [PMID: 27613132 DOI: 10.1016/bs.acr.2016.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metastatic disease is the major cause of fatalities in cancer patients, but few therapies are designed to target the metastatic process. Cancer cells must perform a number of steps to successfully establish metastatic foci, including local invasion, intravasation, survival, extravasation, and growth in ectopic tissue. Due to the nonrandom distribution of metastasis, it has long been recognized that the tissue microenvironment must be an important determinant of colonization. More recently it has been established in animal models that immune cells regulate the metastatic process, including a dominant role for monocytes and macrophages, and emerging roles for neutrophils and various lymphocyte populations. While most research has focused on the early dissemination process, patients usually present clinically with disseminated, if not macroscopic, disease. Identifying pathways by which immune cells regulate growth and therapeutic resistance within metastatic sites is therefore key to the development of pharmacological agents that will significantly extend patient survival.
Collapse
Affiliation(s)
- A de Mingo Pulido
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - B Ruffell
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.
| |
Collapse
|
342
|
McArdle S, Mikulski Z, Ley K. Live cell imaging to understand monocyte, macrophage, and dendritic cell function in atherosclerosis. J Exp Med 2016; 213:1117-31. [PMID: 27270892 PMCID: PMC4925021 DOI: 10.1084/jem.20151885] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/28/2016] [Indexed: 02/06/2023] Open
Abstract
Ley et al. provide a review of the technology and accomplishments of dynamic imaging of myeloid cells in atherosclerosis. Intravital imaging is an invaluable tool for understanding the function of cells in healthy and diseased tissues. It provides a window into dynamic processes that cannot be studied by other techniques. This review will cover the benefits and limitations of various techniques for labeling and imaging myeloid cells, with a special focus on imaging cells in atherosclerotic arteries. Although intravital imaging is a powerful tool for understanding cell function, it alone does not provide a complete picture of the cell. Other techniques, such as flow cytometry and transcriptomics, must be combined with intravital imaging to fully understand a cell's phenotype, lineage, and function.
Collapse
Affiliation(s)
- Sara McArdle
- Division of Inflammation Biology and Microscopy Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA 92037
| | - Zbigniew Mikulski
- Division of Inflammation Biology and Microscopy Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA 92037
| | - Klaus Ley
- Division of Inflammation Biology and Microscopy Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA 92037
| |
Collapse
|
343
|
Chin AR, Wang SE. Cancer Tills the Premetastatic Field: Mechanistic Basis and Clinical Implications. Clin Cancer Res 2016; 22:3725-33. [PMID: 27252414 DOI: 10.1158/1078-0432.ccr-16-0028] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023]
Abstract
A growing body of work has shown that cancer metastasis is not a random spontaneous event; rather, it is the culmination of a cascade of priming steps through which a subpopulation of the tumor cells acquires invasive traits while readying a permissive environment, termed the "premetastatic niche," in which distant metastases can occur. Signals from the primary tumor mobilize and adapt immune cells as well as directly communicating with distant niche cells to induce a broad spectrum of adaptations in target organs, including the induction of angiogenesis, inflammation, extracellular matrix remodeling, and metabolic reprogramming. Together, these interactions facilitate the formation of a premetastatic niche composed of a variable mix of resident and recruited immune cells, endothelial cells, and stromal cells connected through a complex signaling network that we are only beginning to understand. Here, we summarize the latest findings on how cancer induces and guides the formation of this premetastatic niche as well as potential prognostic markers and therapeutic targets that may lead to a better understanding and effective treatment of metastatic disease. Clin Cancer Res; 22(15); 3725-33. ©2016 AACR.
Collapse
Affiliation(s)
- Andrew R Chin
- Department of Cancer Biology, City of Hope Beckman Research Institute, Duarte, California. City of Hope Irell & Manella Graduate School of Biological Sciences, Duarte, California
| | - Shizhen Emily Wang
- Department of Cancer Biology, City of Hope Beckman Research Institute, Duarte, California.
| |
Collapse
|
344
|
Parallel Aspects of the Microenvironment in Cancer and Autoimmune Disease. Mediators Inflamm 2016; 2016:4375120. [PMID: 26997761 PMCID: PMC4779817 DOI: 10.1155/2016/4375120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
Cancer and autoimmune diseases are fundamentally different pathological conditions. In cancer, the immune response is suppressed and unable to eradicate the transformed self-cells, while in autoimmune diseases it is hyperactivated against a self-antigen, leading to tissue injury. Yet, mechanistically, similarities in the triggering of the immune responses can be observed. In this review, we highlight some parallel aspects of the microenvironment in cancer and autoimmune diseases, especially hypoxia, and the role of macrophages, neutrophils, and their interaction. Macrophages, owing to their plastic mode of activation, can generate a pro- or antitumoral microenvironment. Similarly, in autoimmune diseases, macrophages tip the Th1/Th2 balance via various effector cytokines. The contribution of neutrophils, an additional plastic innate immune cell population, to the microenvironment and disease progression is recently gaining more prominence in both cancer and autoimmune diseases, as they can secrete cytokines, chemokines, and reactive oxygen species (ROS), as well as acquire an enhanced ability to produce neutrophil extracellular traps (NETs) that are now considered important initiators of autoimmune diseases. Understanding the contribution of macrophages and neutrophils to the cancerous or autoimmune microenvironment, as well as the role their interaction and cooperation play, may help identify new targets and improve therapeutic strategies.
Collapse
|
345
|
Hanna RN, Chodaczek G, Hedrick CC. In vivo Imaging of Tumor and Immune Cell Interactions in the Lung. Bio Protoc 2016. [PMID: 29516025 DOI: 10.21769/bioprotoc.1973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Immunotherapy has demonstrated great therapeutic potential by activating the immune system to fight cancer. However, little is known about the specific dynamics of interactions that occur between tumor and immune cells. In this protocol we describe a novel method to visualize the interaction of tumor and immune cells in the lung of live mice, which can be applied to other organs. In this protocol fluorescent-labeled tumor cells are transferred to recipient mice expressing fluorescently tagged immune cells. Tumor-immune cell interactions in the lung are then imaged by confocal or two photon microscopy. Analysis of tumor interactions with immune cells using this protocol should aid in a better understanding of the importance of these interactions and their role in developing immunotherapies.
Collapse
Affiliation(s)
- Richard N Hanna
- Department of Respiratory, Inflammation and Autoimmune Diseases, MedImmune, LLC, Gaithersburg, USA.,Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, USA
| | - Grzegorz Chodaczek
- Microscopy Core, La Jolla Institute for Allergy and Immunology, La Jolla, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, USA
| |
Collapse
|
346
|
Abstract
Classical inflammatory monocytes and their derivative macrophages promote tumor metastasis whereas CD8(+) T and NK cells restrict tumor growth. In a recent paper published in Science, Hanna and colleagues demonstrate that another monocyte population, nonclassical patrolling monocytes, is enriched in the microvasculature of tumor-challenged lung and reduces tumor metastasis by recruiting NK cells.
Collapse
|