151
|
Veiseh O, Doloff JC, Ma M, Vegas AJ, Tam HH, Bader AR, Li J, Langan E, Wyckoff J, Loo WS, Jhunjhunwala S, Chiu A, Siebert S, Tang K, Hollister-Lock J, Aresta-Dasilva S, Bochenek M, Mendoza-Elias J, Wang Y, Qi M, Lavin DM, Chen M, Dholakia N, Thakrar R, Lacík I, Weir GC, Oberholzer J, Greiner DL, Langer R, Anderson DG. Size- and shape-dependent foreign body immune response to materials implanted in rodents and non-human primates. NATURE MATERIALS 2015; 14:643-51. [PMID: 25985456 PMCID: PMC4477281 DOI: 10.1038/nmat4290] [Citation(s) in RCA: 611] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/10/2015] [Indexed: 04/14/2023]
Abstract
The efficacy of implanted biomedical devices is often compromised by host recognition and subsequent foreign body responses. Here, we demonstrate the role of the geometry of implanted materials on their biocompatibility in vivo. In rodent and non-human primate animal models, implanted spheres 1.5 mm and above in diameter across a broad spectrum of materials, including hydrogels, ceramics, metals and plastics, significantly abrogated foreign body reactions and fibrosis when compared with smaller spheres. We also show that for encapsulated rat pancreatic islet cells transplanted into streptozotocin-treated diabetic C57BL/6 mice, islets prepared in 1.5-mm alginate capsules were able to restore blood-glucose control for up to 180 days, a period more than five times longer than for transplanted grafts encapsulated within conventionally sized 0.5-mm alginate capsules. Our findings suggest that the in vivo biocompatibility of biomedical devices can be significantly improved simply by tuning their spherical dimensions.
Collapse
Affiliation(s)
- Omid Veiseh
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Joshua C. Doloff
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Minglin Ma
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Arturo J. Vegas
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Hok Hei Tam
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Andrew R. Bader
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Jie Li
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Erin Langan
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Jeffrey Wyckoff
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
| | - Whitney S. Loo
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Siddharth Jhunjhunwala
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Alan Chiu
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Sean Siebert
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Katherine Tang
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Jennifer Hollister-Lock
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | - Stephanie Aresta-Dasilva
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Matthew Bochenek
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Joshua Mendoza-Elias
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Yong Wang
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Merigeng Qi
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Danya M. Lavin
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Michael Chen
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Nimit Dholakia
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Raj Thakrar
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Igor Lacík
- Department for Biomaterials Research, Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia
| | - Gordon C. Weir
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA
| | - Jose Oberholzer
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Dale L. Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Daniel G. Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
152
|
Affiliation(s)
- Ruud A Bank
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
153
|
Abstract
The fate of both endogenous and transplanted stem cells is dependent on the functional status of the regulatory local microenvironment, which is compromised by disease and therapeutic intervention. The glycosaminoglycan hyaluronan (HA) is a critical component of the hematopoietic microenvironment. We summarize recent advances in our understanding of the role of HA in regulating mesenchymal stem cells, osteoblasts, fibroblasts, macrophages, and endothelium in bone marrow (BM) and their crosstalk within the hematopoietic microenvironment. HA not only determines the volume, hydration, and microfluidics of the BM interstitial space, but also, via interactions with specific receptors, regulates multiple cell functions including differentiation, migration, and production of regulatory factors. The effects of HA are dependent on the polymer size and are influenced by the formation of complexes with other molecules. In healthy BM, HA synthases and hyaluronidases form a molecular network that maintains extracellular HA levels within a discrete physiological window, but HA homeostasis is often perturbed in pathological conditions, including hematological malignancies. Recent studies have suggested that HA synthases may have functions beyond HA production and contribute to the intracellular regulatory machinery. We discuss a possible role for HA synthases, intracellular and extracellular HA in the malignant BM microenvironment, and resistance to therapy.
Collapse
|
154
|
Aikawa Y, Yamagata K, Katsumoto T, Shima Y, Shino M, Stanley ER, Cleary ML, Akashi K, Tenen DG, Kitabayashi I. Essential role of PU.1 in maintenance of mixed lineage leukemia-associated leukemic stem cells. Cancer Sci 2015; 106:227-36. [PMID: 25529853 PMCID: PMC4373983 DOI: 10.1111/cas.12593] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/05/2014] [Accepted: 12/14/2014] [Indexed: 01/24/2023] Open
Abstract
Acute myeloid leukemia is a clonal malignant disorder derived from a small number of leukemic stem cells (LSCs). Rearrangements of the mixed lineage leukemia (MLL) gene are found in acute myeloid leukemia associated with poor prognosis. The upregulation of Hox genes is critical for LSC induction and maintenance, but is unlikely to support malignancy and the high LSC frequency observed in MLL leukemias. The present study shows that MLL fusion proteins interact with the transcription factor PU.1 to activate the transcription of CSF-1R, which is critical for LSC activity. Acute myeloid leukemia is cured by either deletion of PU.1 or ablation of cells expressing CSF-1R. Kinase inhibitors specific for CSF-1R prolong survival time. These findings indicate that PU.1-mediated upregulation of CSF-1R is a critical effector of MLL leukemogenesis.
Collapse
Affiliation(s)
- Yukiko Aikawa
- Division of Hematological Malignancy, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Naik S, Bouladoux N, Linehan JL, Han SJ, Harrison OJ, Wilhelm C, Conlan S, Himmelfarb S, Byrd AL, Deming C, Quinones M, Brenchley JM, Kong HH, Tussiwand R, Murphy KM, Merad M, Segre JA, Belkaid Y. Commensal-dendritic-cell interaction specifies a unique protective skin immune signature. Nature 2015; 520:104-8. [PMID: 25539086 DOI: 10.1038/nature14052] [Citation(s) in RCA: 594] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023]
Abstract
The skin represents the primary interface between the host and the environment. This organ is also home to trillions of microorganisms that play an important role in tissue homeostasis and local immunity. Skin microbial communities are highly diverse and can be remodelled over time or in response to environmental challenges. How, in the context of this complexity, individual commensal microorganisms may differentially modulate skin immunity and the consequences of these responses for tissue physiology remains unclear. Here we show that defined commensals dominantly affect skin immunity and identify the cellular mediators involved in this specification. In particular, colonization with Staphylococcus epidermidis induces IL-17A(+) CD8(+) T cells that home to the epidermis, enhance innate barrier immunity and limit pathogen invasion. Commensal-specific T-cell responses result from the coordinated action of skin-resident dendritic cell subsets and are not associated with inflammation, revealing that tissue-resident cells are poised to sense and respond to alterations in microbial communities. This interaction may represent an evolutionary means by which the skin immune system uses fluctuating commensal signals to calibrate barrier immunity and provide heterologous protection against invasive pathogens. These findings reveal that the skin immune landscape is a highly dynamic environment that can be rapidly and specifically remodelled by encounters with defined commensals, findings that have profound implications for our understanding of tissue-specific immunity and pathologies.
Collapse
Affiliation(s)
- Shruti Naik
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Nicolas Bouladoux
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Jonathan L Linehan
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Seong-Ji Han
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Oliver J Harrison
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Christoph Wilhelm
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Sean Conlan
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Sarah Himmelfarb
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| | - Allyson L Byrd
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA [3] Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Clayton Deming
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Mariam Quinones
- Bioinformatics and Computational Bioscience Branch, National Institute of Allergy and Infectious Diseases, NIH Bethesda, Maryland 20892, USA
| | - Jason M Brenchley
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Immunopathogenesis Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, NIH Bethesda, Maryland 20892, USA
| | - Heidi H Kong
- Dermatology Branch, National Cancer Institute, NIH Bethesda, Maryland 20892, USA
| | - Roxanne Tussiwand
- Howard Hughes Medical Institute, Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Kenneth M Murphy
- Howard Hughes Medical Institute, Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Julia A Segre
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Yasmine Belkaid
- 1] Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA [2] Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA
| |
Collapse
|
156
|
Patel SK, Janjic JM. Macrophage targeted theranostics as personalized nanomedicine strategies for inflammatory diseases. Am J Cancer Res 2015; 5:150-72. [PMID: 25553105 PMCID: PMC4279001 DOI: 10.7150/thno.9476] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/28/2014] [Indexed: 12/16/2022] Open
Abstract
Inflammatory disease management poses challenges due to the complexity of inflammation and inherent patient variability, thereby necessitating patient-specific therapeutic interventions. Theranostics, which integrate therapeutic and imaging functionalities, can be used for simultaneous imaging and treatment of inflammatory diseases. Theranostics could facilitate assessment of safety, toxicity and real-time therapeutic efficacy leading to personalized treatment strategies. Macrophages are an important cellular component of inflammatory diseases, participating in varied roles of disease exacerbation and resolution. The inherent phagocytic nature, abundance and disease homing properties of macrophages can be targeted for imaging and therapeutic purposes. This review discusses the utility of theranostics in macrophage ablation, phenotype modulation and inhibition of their inflammatory activity leading to resolution of inflammation in several diseases.
Collapse
|
157
|
Blériot C, Dupuis T, Jouvion G, Eberl G, Disson O, Lecuit M. Liver-resident macrophage necroptosis orchestrates type 1 microbicidal inflammation and type-2-mediated tissue repair during bacterial infection. Immunity 2014; 42:145-58. [PMID: 25577440 DOI: 10.1016/j.immuni.2014.12.020] [Citation(s) in RCA: 357] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/01/2014] [Accepted: 11/26/2014] [Indexed: 12/18/2022]
Abstract
Kupffer cells, the phagocytes of fetal origin that line the liver sinusoids, are key contributors of host defense against enteroinvasive bacteria. Here, we found that infection by Listeria monocytogenes induced the early necroptotic death of Kupffer cells, which was followed by monocyte recruitment and an anti-bacterial type 1 inflammatory response. Kupffer cell death also triggered a type 2 response that involved the hepatocyte-derived alarmin interleukin-33 (IL-33) and basophil-derived interleukin-4 (IL-4). This led to the alternative activation of the monocyte-derived macrophages recruited to the liver, which thereby replaced ablated Kupffer cells and restored liver homeostasis. Kupffer cell death is therefore a key signal orchestrating type 1 microbicidal inflammation and type-2-mediated liver repair upon infection. This indicates that beyond the classical dichotomy of type 1 and type 2 responses, these responses can develop sequentially in the context of a bacterial infection and act interdependently, orchestrating liver immune responses and return to homeostasis, respectively.
Collapse
Affiliation(s)
- Camille Blériot
- Institut Pasteur, Biology of Infection Unit, 75015 Paris, France; Inserm U1117, 75015 Paris, France
| | - Théo Dupuis
- Institut Pasteur, Biology of Infection Unit, 75015 Paris, France; Inserm U1117, 75015 Paris, France
| | - Grégory Jouvion
- Institut Pasteur, Human Histopathology and Animal Models Unit, 75015 Paris, France
| | - Gérard Eberl
- Institut Pasteur, Lymphoid Tissue Development Unit, 75015 Paris, France
| | - Olivier Disson
- Institut Pasteur, Biology of Infection Unit, 75015 Paris, France; Inserm U1117, 75015 Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, 75015 Paris, France; Inserm U1117, 75015 Paris, France; Institut Pasteur, French National Reference Center and World Health Organization Collaborating Centre on Listeria, 75015 Paris, France; Paris Descartes University, Sorbonne Paris Cité, Institut Imagine, Division of Infectious Diseases and Tropical Medicine, Necker-Pasteur Centre for Infectiology, Necker-Enfants Malades University Hospital, 75015 Paris, France.
| |
Collapse
|
158
|
Evrard M, Chong SZ, Devi S, Chew WK, Lee B, Poidinger M, Ginhoux F, Tan SM, Ng LG. Visualization of bone marrow monocyte mobilization using Cx3cr1gfp/+Flt3L-/- reporter mouse by multiphoton intravital microscopy. J Leukoc Biol 2014; 97:611-9. [PMID: 25516753 DOI: 10.1189/jlb.1ta0514-274r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Monocytes are innate immune cells that play critical roles in inflammation and immune defense. A better comprehension of how monocytes are mobilized and recruited is fundamental to understand their biologic role in disease and steady state. The BM represents a major "checkpoint" for monocyte homeostasis, as it is the primary site for their production and release. Our study determined that the Cx3cr1(gfp/+) mouse strain is currently the most ideal model for the visualization of monocyte behavior in the BM by multiphoton intravital microscopy. However, we observed that DCs are also labeled with high levels of GFP and thus, interfere with the accuracy of monocyte tracking in vivo. Hence, we generated a Cx3cr1(gfp/+)Flt3L(-/-) reporter mouse and showed that whereas monocyte numbers were not affected, DC numbers were reduced significantly, as DCs but not monocytes depend on Flt3 signaling for their development. We thus verified that mobilization of monocytes from the BM in Cx3cr1(gfp/+)Flt3L(-/-) mice is intact in response to LPS. Collectively, our study demonstrates that the Cx3cr1(gfp/+)Flt3L(-/-) reporter mouse model represents a powerful tool to visualize monocyte activities in BM and illustrates the potential of a Cx3cr1(gfp/+)-based, multifunctionality fluorescence reporter approach to dissect monocyte function in vivo.
Collapse
Affiliation(s)
- Maximilien Evrard
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Shu Zhen Chong
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Sapna Devi
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Weng Keong Chew
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Bernett Lee
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Michael Poidinger
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Florent Ginhoux
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Suet Mien Tan
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Lai Guan Ng
- *Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; and School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
159
|
Impaired eukaryotic translation initiation factor 2B activity specifically in oligodendrocytes reproduces the pathology of vanishing white matter disease in mice. J Neurosci 2014; 34:12182-91. [PMID: 25186761 DOI: 10.1523/jneurosci.1373-14.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vanishing white matter disease (VWMD) is an inherited autosomal-recessive hypomyelinating disease caused by mutations in eukaryotic translation initiation factor 2B (eIF2B). eIF2B mutations predominantly affect the brain white matter, and the characteristic features of VWMD pathology include myelin loss and foamy oligodendrocytes. Activation of pancreatic endoplasmic reticulum kinase (PERK) has been observed in oligodendrocytes in VWMD. PERK activation in response to endoplasmic reticulum stress attenuates eIF2B activity by phosphorylating eIF2α, suggesting that impaired eIF2B activity in oligodendrocytes induced by VWMD mutations or PERK activation exploit similar mechanisms to promote selective white matter pathology in VWMD. Using transgenic mice that allow for temporally controlled activation of PERK specifically in oligodendrocytes, we discovered that strong PERK activation in oligodendrocytes during development suppressed eIF2B activity and reproduced the characteristic features of VWMD in mice, including hypomyelinating phenotype, foamy oligodendrocytes, and myelin loss. Notably, impaired eIF2B activity induced by PERK activation in oligodendrocytes of fully myelinated adult mice had minimal effects on morphology or function. Our observations point to a cell-autonomous role of impaired eIF2B activity in myelinating oligodendrocytes in the pathogenesis of VWMD.
Collapse
|
160
|
Ye Z, Gorman AA, Uittenbogaard AM, Myers-Morales T, Kaplan AM, Cohen DA, Straley SC. Caspase-3 mediates the pathogenic effect of Yersinia pestis YopM in liver of C57BL/6 mice and contributes to YopM's function in spleen. PLoS One 2014; 9:e110956. [PMID: 25372388 PMCID: PMC4220956 DOI: 10.1371/journal.pone.0110956] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 09/26/2014] [Indexed: 12/15/2022] Open
Abstract
The virulence protein YopM of the plague bacterium Yersinia pestis has different dominant effects in liver and spleen. Previous studies focused on spleen, where YopM inhibits accumulation of inflammatory dendritic cells. In the present study we focused on liver, where PMN function may be directly undermined by YopM without changes in inflammatory cell numbers in the initial days of infection, and foci of inflammation are easily identified. Mice were infected with parent and ΔyopM-1 Y. pestis KIM5, and effects of YopM were assessed by immunohistochemistry and determinations of bacterial viable numbers in organs. The bacteria were found associated with myeloid cells in foci of inflammation and in liver sinusoids. A new in-vivo phenotype of YopM was revealed: death of inflammatory cells, evidenced by TUNEL staining beginning at d 1 of infection. Based on distributions of Ly6G+, F4/80+, and iNOS+ cells within foci, the cells that were killed could have included both PMNs and macrophages. By 2 d post-infection, YopM had no effect on distribution of these cells, but by 3 d cellular decomposition had outstripped acute inflammation in foci due to parent Y. pestis, while foci due to the ΔyopM-1 strain still contained many inflammatory cells. The destruction depended on the presence of both PMNs in the mice and YopM in the bacteria. In mice that lacked the apoptosis mediator caspase-3 the infection dynamics were novel: the parent Y. pestis was limited in growth comparably to the ΔyopM-1 strain in liver, and in spleen a partial growth limitation for parent Y. pestis was seen. This result identified caspase-3 as a co-factor or effector in YopM's action and supports the hypothesis that in liver YopM's main pathogenic effect is mediated by caspase-3 to cause apoptosis of PMNs.
Collapse
Affiliation(s)
- Zhan Ye
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
| | - Amanda A. Gorman
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
| | - Annette M. Uittenbogaard
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
| | - Tanya Myers-Morales
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
| | - Alan M. Kaplan
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
| | - Donald A. Cohen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
| | - Susan C. Straley
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States of America
- * E-mail:
| |
Collapse
|
161
|
Raggatt LJ, Wullschleger ME, Alexander KA, Wu ACK, Millard SM, Kaur S, Maugham ML, Gregory LS, Steck R, Pettit AR. Fracture healing via periosteal callus formation requires macrophages for both initiation and progression of early endochondral ossification. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3192-204. [PMID: 25285719 DOI: 10.1016/j.ajpath.2014.08.017] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 08/18/2014] [Accepted: 08/21/2014] [Indexed: 11/29/2022]
Abstract
The distribution, phenotype, and requirement of macrophages for fracture-associated inflammation and/or early anabolic progression during endochondral callus formation were investigated. A murine femoral fracture model [internally fixed using a flexible plate (MouseFix)] was used to facilitate reproducible fracture reduction. IHC demonstrated that inflammatory macrophages (F4/80(+)Mac-2(+)) were localized with initiating chondrification centers and persisted within granulation tissue at the expanding soft callus front. They were also associated with key events during soft-to-hard callus transition. Resident macrophages (F4/80(+)Mac-2(neg)), including osteal macrophages, predominated in the maturing hard callus. Macrophage Fas-induced apoptosis transgenic mice were used to induce macrophage depletion in vivo in the femoral fracture model. Callus formation was completely abolished when macrophage depletion was initiated at the time of surgery and was significantly reduced when depletion was delayed to coincide with initiation of early anabolic phase. Treatment initiating 5 days after fracture with the pro-macrophage cytokine colony stimulating factor-1 significantly enhanced soft callus formation. The data support that inflammatory macrophages were required for initiation of fracture repair, whereas both inflammatory and resident macrophages promoted anabolic mechanisms during endochondral callus formation. Overall, macrophages make substantive and prolonged contributions to fracture healing and can be targeted as a therapeutic approach for enhancing repair mechanisms. Thus, macrophages represent a viable target for the development of pro-anabolic fracture treatments with a potentially broad therapeutic window.
Collapse
Affiliation(s)
- Liza J Raggatt
- Bone and Immunology Laboratory, Mater Research Institute-UQ, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia; UQ-Centre for Clinical Research, Faculty of Health Sciences, The University of Queensland, Herston, Queensland, Australia
| | - Martin E Wullschleger
- Trauma Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; School of Medicine, Faculty of Health Sciences, The University of Queensland, Herston, Queensland, Australia
| | - Kylie A Alexander
- UQ-Centre for Clinical Research, Faculty of Health Sciences, The University of Queensland, Herston, Queensland, Australia
| | - Andy C K Wu
- UQ-Centre for Clinical Research, Faculty of Health Sciences, The University of Queensland, Herston, Queensland, Australia
| | - Susan M Millard
- Bone and Immunology Laboratory, Mater Research Institute-UQ, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Simranpreet Kaur
- Bone and Immunology Laboratory, Mater Research Institute-UQ, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia; UQ-Centre for Clinical Research, Faculty of Health Sciences, The University of Queensland, Herston, Queensland, Australia
| | - Michelle L Maugham
- Bone and Immunology Laboratory, Mater Research Institute-UQ, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Laura S Gregory
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Roland Steck
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Allison R Pettit
- Bone and Immunology Laboratory, Mater Research Institute-UQ, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia; UQ-Centre for Clinical Research, Faculty of Health Sciences, The University of Queensland, Herston, Queensland, Australia.
| |
Collapse
|
162
|
Heipertz EL, Davies ML, Lin E, Norbury CC. Prolonged antigen presentation following an acute virus infection requires direct and then cross-presentation. THE JOURNAL OF IMMUNOLOGY 2014; 193:4169-77. [PMID: 25225666 DOI: 10.4049/jimmunol.1302565] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Antiviral CD8(+) T cell recognition of MHC class I-peptide complexes on the surface of professional APCs is a requisite step in an effective immune response following many potentially lethal infections. Although MHC class I-peptide production is thought to be closely linked to the continued presence of virus, several studies have shown that the persistence of Ag presentation occurs for an extended period of time following the clearance of RNA viruses. However, the mechanism responsible for Ag presentation persistence following viral clearance was unknown until now. In this study, we used a recombinant DNA virus expressing different forms of a model Ag to study the mechanism of prolonged Ag presentation in mice. We determined that the persistence of Ag presentation consists of three distinct mechanistic phases, as follows: ongoing viral replication, persistence of virally infected cells, and cross-presentation of Ag. These data will allow manipulation of the form of Ag contained within viral vectors to produce the most effective and protective CD8(+) T cell response to be generated following vaccination.
Collapse
Affiliation(s)
- Erica L Heipertz
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033
| | - Michael L Davies
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033
| | - Eugene Lin
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033
| | - Christopher C Norbury
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033
| |
Collapse
|
163
|
Stouch AN, Zaynagetdinov R, Barham WJ, Stinnett AM, Slaughter JC, Yull FE, Hoffman HM, Blackwell TS, Prince LS. IκB kinase activity drives fetal lung macrophage maturation along a non-M1/M2 paradigm. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:1184-93. [PMID: 24981452 PMCID: PMC4108541 DOI: 10.4049/jimmunol.1302516] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In preterm infants, exposure to inflammation increases the risk of bronchopulmonary dysplasia, a chronic, developmental lung disease. Although macrophages are the key cells that initiate lung inflammation, less is known about lung macrophage phenotype and maturation. We hypothesized that fetal lung macrophages mature into distinct subpopulations during mouse development, and that activation could influence macrophage maturation. Expression of the fetal macrophage markers CD68, CD86, CD206, Ym1, fibrinogen-like protein 2, and indolamine-2, 3-dioxygenase was developmentally regulated, with each marker having different temporal patterns. Flow cytometry analysis showed macrophages within the fetal lung were less diverse than the distinctly separate subpopulations in newborn and adult lungs. Similar to adult alveolar macrophages, fetal lung macrophages responded to the TLR4 agonist LPS and the alternative activation cytokines IL-4 and IL-13. Using a macrophage-specific constitutively active IκB Kinase transgenic model (IKFM), we demonstrated that macrophage activation increased proinflammatory gene expression and reduced the response of fetal lung macrophages to IL-4 and IL-13. Activation also increased fetal lung macrophage proliferation. Fetal IKFM lungs contained increased percentages of more mature, CD11b(low)F4/80(high) cells that also expressed higher levels of the alternative activation markers CD204 and CD206. Development of fetal lung macrophages into mature alveolar macrophages may therefore include features of both proinflammatory and alternative activation paradigms.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Biomarkers/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Enzyme Activation/immunology
- Female
- Gene Expression Regulation, Developmental/immunology
- Gene Expression Regulation, Enzymologic/immunology
- Humans
- I-kappa B Kinase/metabolism
- I-kappa B Kinase/physiology
- Immunophenotyping
- Inflammation/enzymology
- Inflammation/immunology
- Inflammation/pathology
- Lung Diseases/enzymology
- Lung Diseases/immunology
- Lung Diseases/pathology
- Macrophage Activation/immunology
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/pathology
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
Collapse
Affiliation(s)
- Ashley N Stouch
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Rinat Zaynagetdinov
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Whitney J Barham
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Amanda M Stinnett
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - James C Slaughter
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Fiona E Yull
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Hal M Hoffman
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Timothy S Blackwell
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| | - Lawrence S Prince
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093; Rady Children's Hospital, San Diego, CA 92123;Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232;Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; andDepartment of Biostatistics, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
164
|
Macrophages in cardiac homeostasis, injury responses and progenitor cell mobilisation. Stem Cell Res 2014; 13:705-14. [PMID: 25087895 DOI: 10.1016/j.scr.2014.06.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 06/26/2014] [Indexed: 12/23/2022] Open
Abstract
Macrophages are an immune cell type found in every organ of the body. Classically, macrophages are recognised as housekeeping cells involved in the detection of foreign antigens and danger signatures, and the clearance of tissue debris. However, macrophages are increasingly recognised as a highly versatile cell type with a diverse range of functions that are important for tissue homeostasis and injury responses. Recent research findings suggest that macrophages contribute to tissue regeneration and may play a role in the activation and mobilisation of stem cells. This review describes recent advances in our understanding of the role played by macrophages in cardiac tissue maintenance and repair following injury. We examine the involvement of exogenous and resident tissue macrophages in cardiac inflammatory responses and their potential activity in regulating cardiac regeneration.
Collapse
|
165
|
Yang L, Kwon J, Popov Y, Gajdos GB, Ordog T, Brekken RA, Mukhopadhyay D, Schuppan D, Bi Y, Simonetto D, Shah VH. Vascular endothelial growth factor promotes fibrosis resolution and repair in mice. Gastroenterology 2014; 146:1339-50.e1. [PMID: 24503129 PMCID: PMC4001704 DOI: 10.1053/j.gastro.2014.01.061] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 01/20/2014] [Accepted: 01/29/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Vascular endothelial growth factor (VEGF)-induced angiogenesis is implicated in fibrogenesis and portal hypertension. However, the function of VEGF in fibrosis resolution has not been explored. METHODS We developed a cholecystojejunostomy procedure to reconstruct biliary flow after bile duct ligation in C57BL/6 mice to generate a model of fibrosis resolution. These mice were then given injections of VEGF-neutralizing (mcr84) or control antibodies, and other mice received an adenovirus that expressed mouse VEGF or a control vector. The procedure was also performed on macrophage fas-induced apoptosis mice, in which macrophages can be selectively depleted. Liver and blood samples were collected and analyzed in immunohistochemical, morphometric, vascular permeability, real-time polymerase chain reaction, and flow cytometry assays. RESULTS VEGF-neutralizing antibodies prevented development of fibrosis but also disrupted hepatic tissue repair and fibrosis resolution. During fibrosis resolution, VEGF inhibition impaired liver sinusoidal permeability, which was associated with reduced monocyte migration, adhesion, and infiltration of fibrotic liver. Scar-associated macrophages contributed to this process by producing the chemokine (C-X-C motif) ligand 9 (CXCL9) and matrix metalloproteinase 13. Resolution of fibrosis was impaired in macrophage fas-induced apoptosis mice but increased after overexpression of CXCL9. CONCLUSIONS In a mouse model of liver fibrosis resolution, VEGF promoted fibrogenesis, but was also required for hepatic tissue repair and fibrosis resolution. We observed that VEGF regulates vascular permeability, monocyte infiltration, and scar-associated macrophages function.
Collapse
Affiliation(s)
- Liu Yang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Junghee Kwon
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Yury Popov
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Gabriella B. Gajdos
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Tamas Ordog
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, Division of Surgical Oncology, Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | | | - Detlef Schuppan
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Yan Bi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Douglas Simonetto
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
166
|
Redente EF, Keith RC, Janssen W, Henson PM, Ortiz LA, Downey GP, Bratton DL, Riches DWH. Tumor necrosis factor-α accelerates the resolution of established pulmonary fibrosis in mice by targeting profibrotic lung macrophages. Am J Respir Cell Mol Biol 2014; 50:825-37. [PMID: 24325577 PMCID: PMC4068926 DOI: 10.1165/rcmb.2013-0386oc] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/04/2013] [Indexed: 01/13/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which alternatively programmed macrophages produce profibrotic molecules that promote myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF are lacking, and conventional therapy may be harmful. We tested the hypothesis that therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of established pulmonary fibrosis. Fibrosis was assessed in bleomycin-instilled wild-type and TNF-α(-/-) mice by measuring hydroxyproline levels, static compliance, and Masson's trichrome staining. Macrophage infiltration and programming status was assessed by flow cytometry of enzymatically digested lung and in situ immunostaining. Pulmonary delivery of TNF-α to wild-type mice with established pulmonary fibrosis was found to reduce their fibrotic burden, to improve lung function and architecture, and to reduce the number and programming status of profibrotic alternatively programmed macrophages. In contrast, fibrosis and alternative macrophage programming were prolonged in bleomycin-instilled TNF-α(-/-) mice. To address the role of the reduced numbers of alternatively programmed macrophages in the TNF-α-induced resolution of established pulmonary fibrosis, we conditionally depleted macrophages in MAFIA (MAcrophage Fas-Induced Apoptosis) mice. Conditional macrophage depletion phenocopied the resolution of established pulmonary fibrosis observed after therapeutic TNF-α delivery. Taken together, our results show for the first time that TNF-α is involved in the resolution of established pulmonary fibrosis via a mechanism involving reduced numbers and programming status of profibrotic macrophages. We speculate that pulmonary delivery of TNF-α or augmenting its signaling pathway represent a novel therapeutic strategy to resolve established pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Rebecca C. Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
| | - William Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
| | - Peter M. Henson
- Program in Cell Biology, Department of Pediatrics, and
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
| | - Luis A. Ortiz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh Pennsylvania
| | - Gregory P. Downey
- Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
| | | | - David W. H. Riches
- Program in Cell Biology, Department of Pediatrics, and
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado; and
| |
Collapse
|
167
|
He H, Tan Y, Duffort S, Perez VL, Tseng SCG. In vivo downregulation of innate and adaptive immune responses in corneal allograft rejection by HC-HA/PTX3 complex purified from amniotic membrane. Invest Ophthalmol Vis Sci 2014; 55:1647-56. [PMID: 24519420 DOI: 10.1167/iovs.13-13094] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Heavy chain-hyaluronic acid (HC-HA)/PTX3 purified from human amniotic membrane (AM) was previously observed to suppress inflammatory responses in vitro. We now examine whether HC-HA/PTX3 is able to exert a similar effect in vivo, using murine models for keratitis and corneal allograft rejection. METHODS The in vitro effect of HC-HA/PTX3 was tested using OTII ovalbumin (OVA) transgenic, purified CD4(+) T cells, or IFN-γ/lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Cytokine production was measured by ELISA, while cell surface markers and cell proliferation were determined by flow cytometry. In vivo effects of HC-HA/PTX3 were analyzed by quantifying the recruitment of enhanced green fluorescence-labeled macrophages and by measuring the expression of arginase 1 (Arg-1), IL-10, and IL-12 in LPS-induced keratitis in the macrophage Fas-induced apoptosis (Mafia) mouse. The effect of corneal allograft survival in a complete major histocompatibility complex (MHC) mismatched mouse model was assessed by grading corneal opacification. RESULTS In vitro studies demonstrated that HC-HA/PTX3 significantly enhanced the expansion of FOXP3 T cells and suppressed cell proliferation and protein expression of IFN-γ, IL-2, CD25, and CD69 in activated CD4(+) T cells. Furthermore, immobilized HC-HA/PTX3 significantly upregulated IL-10 gene expression but downregulated that of IL-12 and IL-23 in activated RAW264.7 cells. Finally, in vivo subconjunctival injection of HC-HA/PTX3 significantly prolonged corneal allograft survival, suppressed macrophage infiltration, and promoted M2 polarization by upregulating Arg-1 and IL-10 but downregulating IL-12. CONCLUSIONS HC-HA/PTX3 can suppress inflammatory responses in vivo by modulating both innate and adaptive immunity of macrophages and CD4(+) T cells.
Collapse
Affiliation(s)
- Hua He
- TissueTech, Inc., Miami, Florida
| | | | | | | | | |
Collapse
|
168
|
Osteal macrophages support physiologic skeletal remodeling and anabolic actions of parathyroid hormone in bone. Proc Natl Acad Sci U S A 2014; 111:1545-50. [PMID: 24406853 DOI: 10.1073/pnas.1315153111] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cellular subpopulations in the bone marrow play distinct and unexplored functions in skeletal homeostasis. This study delineated a unique role of osteal macrophages in bone and parathyroid hormone (PTH)-dependent bone anabolism using murine models of targeted myeloid-lineage cell ablation. Depletion of c-fms(+) myeloid lineage cells [via administration of AP20187 in the macrophage Fas-induced apoptosis (MAFIA) mouse model] reduced cortical and trabecular bone mass and attenuated PTH-induced trabecular bone anabolism, supporting the positive function of macrophages in bone homeostasis. Interestingly, using a clodronate liposome model with targeted depletion of mature phagocytic macrophages an opposite effect was found with increased trabecular bone mass and increased PTH-induced anabolism. Apoptotic cells were more numerous in MAFIA versus clodronate-treated mice and flow cytometric analyses of myeloid lineage cells in the bone marrow showed that MAFIA mice had reduced CD68(+) cells, whereas clodronate liposome-treated mice had increased CD68(+) and CD163(+) cells. Clodronate liposomes increased efferocytosis (clearance of apoptotic cells) and gene expression associated with alternatively activated M2 macrophages as well as expression of genes associated with bone formation including Wnt3a, Wnt10b, and Tgfb1. Taken together, depletion of early lineage macrophages resulted in osteopenia with blunted effects of PTH anabolic actions, whereas depletion of differentiated macrophages promoted apoptotic cell clearance and transformed the bone marrow to an osteogenic environment with enhanced PTH anabolism. These data highlight a unique function for osteal macrophages in skeletal homeostasis.
Collapse
|
169
|
Abstract
Targeted cell ablation has proven to be a valuable approach to study in vivo cell functions during organogenesis, tissue homeostasis, and regeneration. Over the last two decades, various approaches have been developed to refine the control of cell ablation. In this review, we give an overview of the distinct genetic tools available for targeted cell ablation, with a particular emphasis on their respective specificity.
Collapse
|
170
|
Chèvre R, González-Granado JM, Megens RTA, Sreeramkumar V, Silvestre-Roig C, Molina-Sánchez P, Weber C, Soehnlein O, Hidalgo A, Andrés V. High-resolution imaging of intravascular atherogenic inflammation in live mice. Circ Res 2013; 114:770-9. [PMID: 24366169 DOI: 10.1161/circresaha.114.302590] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE The inflammatory processes that initiate and propagate atherosclerosis remain poorly understood, largely because defining the intravascular behavior of immune cells has been technically challenging. Respiratory and pulsatile movements have hampered in vivo visualization of leukocyte accumulation in athero-prone arteries at resolutions achieved in other tissues. OBJECTIVE To establish and to validate a method that allows high-resolution imaging of inflammatory leukocytes and platelets within the carotid artery of atherosusceptible mice in vivo. METHODS AND RESULTS We have devised a procedure to stabilize the mouse carotid artery mechanically without altering blood dynamics, which dramatically enhances temporal and spatial resolutions using high-speed intravital microscopy in multiple channels of fluorescence. By applying this methodology at different stages of disease progression in atherosusceptible mice, we first validated our approach by assessing the recruitment kinetics of various leukocyte subsets and platelets in athero-prone segments of the carotid artery. The high temporal and spatial resolution allowed the dissection of both the dynamic polarization of and the formation of subcellular domains within adhered leukocytes. We further demonstrate that the secondary capture of activated platelets on the plaque is predominantly mediated by neutrophils. Finally, we couple this procedure with triggered 2-photon microscopy to visualize the 3-dimensional movement of leukocytes in intimate contact with the arterial lumen. CONCLUSIONS The improved imaging of diseased arteries at subcellular resolution presented here should help resolve many outstanding questions in atherosclerosis and other arterial disorders.
Collapse
Affiliation(s)
- Raphael Chèvre
- From the Department of Epidemiology, Atherothrombosis, and Imaging, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.C., J.M.G.-G., V.S., C.S.-R., P.M.-S., A.H., V.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (R.T.A.M., C.W., O.S.); Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands (R.T.A.M., C.W.); and Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (O.S.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Cavnar MJ, Zeng S, Kim TS, Sorenson EC, Ocuin LM, Balachandran VP, Seifert AM, Greer JB, Popow R, Crawley MH, Cohen NA, Green BL, Rossi F, Besmer P, Antonescu CR, DeMatteo RP. KIT oncogene inhibition drives intratumoral macrophage M2 polarization. ACTA ACUST UNITED AC 2013; 210:2873-86. [PMID: 24323358 PMCID: PMC3865475 DOI: 10.1084/jem.20130875] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Imatinib reduces tumor cell KIT signaling and causes tumor cell apoptosis, which drives TAMs to shift from M1- to M2-like in mouse and human GIST. Tumor-associated macrophages (TAMs) are a major component of the cancer microenvironment. Modulation of TAMs is under intense investigation because they are thought to be nearly always of the M2 subtype, which supports tumor growth. Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and typically results from an activating mutation in the KIT oncogene. Using a spontaneous mouse model of GIST and 57 freshly procured human GISTs, we discovered that TAMs displayed an M1-like phenotype and function at baseline. In both mice and humans, the KIT oncoprotein inhibitor imatinib polarized TAMs to become M2-like, a process which involved TAM interaction with apoptotic tumor cells leading to the induction of CCAAT/enhancer binding protein (C/EBP) transcription factors. In human GISTs that eventually developed resistance to imatinib, TAMs reverted to an M1-like phenotype and had a similar gene expression profile as TAMs from untreated human GISTs. Therefore, TAM polarization depends on tumor cell oncogene activity and has important implications for immunotherapeutic strategies in human cancers.
Collapse
Affiliation(s)
- Michael J Cavnar
- Department of Surgery, 2 Department of Developmental Biology, and 3 Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Lin Y, Huang G, Jamison S, Li J, Harding HP, Ron D, Lin W. PERK activation preserves the viability and function of remyelinating oligodendrocytes in immune-mediated demyelinating diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:507-19. [PMID: 24269558 DOI: 10.1016/j.ajpath.2013.10.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/24/2013] [Accepted: 10/23/2013] [Indexed: 10/26/2022]
Abstract
Remyelination occurs in multiple sclerosis (MS) lesions but is generally considered to be insufficient. One of the major challenges in MS research is to understand the causes of remyelination failure and to identify therapeutic targets that promote remyelination. Activation of pancreatic endoplasmic reticulum kinase (PERK) signaling in response to endoplasmic reticulum stress modulates cell viability and function under stressful conditions. There is evidence that PERK is activated in remyelinating oligodendrocytes in demyelinated lesions in both MS and its animal model, experimental autoimmune encephalomyelitis (EAE). In this study, we sought to determine the role of PERK signaling in remyelinating oligodendrocytes in MS and EAE using transgenic mice that allow temporally controlled activation of PERK signaling specifically in oligodendrocytes. We demonstrated that persistent PERK activation was not deleterious to myelinating oligodendrocytes in young, developing mice or to remyelinating oligodendrocytes in cuprizone-induced demyelinated lesions. We found that enhancing PERK activation, specifically in (re)myelinating oligodendrocytes, protected the cells and myelin against the detrimental effects of interferon-γ, a key proinflammatory cytokine in MS and EAE. More important, we showed that enhancing PERK activation in remyelinating oligodendrocytes at the recovery stage of EAE promoted cell survival and remyelination in EAE demyelinated lesions. Thus, our data provide direct evidence that PERK activation cell-autonomously enhances the survival and preserves function of remyelinating oligodendrocytes in immune-mediated demyelinating diseases.
Collapse
Affiliation(s)
- Yifeng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Guangcun Huang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Stephanie Jamison
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Jin Li
- Department of Ophthalmology, 9th Hospital, Shanghai Jiaotong University School of Medical Science, Shanghai, China
| | - Heather P Harding
- University of Cambridge Metabolic Research Laboratories, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - David Ron
- University of Cambridge Metabolic Research Laboratories, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
173
|
Montalvao F, Garcia Z, Celli S, Breart B, Deguine J, Van Rooijen N, Bousso P. The mechanism of anti-CD20-mediated B cell depletion revealed by intravital imaging. J Clin Invest 2013; 123:5098-103. [PMID: 24177426 DOI: 10.1172/jci70972] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/22/2013] [Indexed: 12/14/2022] Open
Abstract
Anti-CD20 Ab therapy has proven successful for treating B cell malignancies and a number of autoimmune diseases. However, how anti-CD20 Abs operate in vivo to mediate B cell depletion is not fully understood. In particular, the anatomical location, the type of effector cells, and the mechanism underlying anti-CD20 therapy remain uncertain. Here, we found that the liver is a major site for B cell depletion and that recirculation accounts for the decrease in B cell numbers observed in secondary lymphoid organs. Using intravital imaging, we established that, upon anti-CD20 treatment, Kupffer cells (KCs) mediate the abrupt arrest and subsequent engulfment of B cells circulating in the liver sinusoids. KCs were also effective in depleting malignant B cells in a model of spontaneous lymphoma. Our results identify Ab-dependent cellular phagocytosis by KCs as a primary mechanism of anti-CD20 therapy and provide an experimental framework for optimizing the efficacy of therapeutic Abs.
Collapse
|
174
|
Wang NK, Lai CC, Liu CH, Yeh LK, Chou CL, Kong J, Nagasaki T, Tsang SH, Chien CL. Origin of fundus hyperautofluorescent spots and their role in retinal degeneration in a mouse model of Goldmann-Favre syndrome. Dis Model Mech 2013; 6:1113-22. [PMID: 23828046 PMCID: PMC3759331 DOI: 10.1242/dmm.012112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Goldmann-Favre syndrome, also known as enhanced S-cone syndrome, is an inherited retinal degeneration disease in which a gain of photoreceptor cell types results in retinal dysplasia and degeneration. Although microglia have been implicated in the pathogenesis of many neurodegenerative diseases, the fundamental role of these cells in this disease is unknown. In the current study, sequential analyses suggest that microglia are recruited and appear after outer nuclear layer folding. By crossing rd7 mice (a model for hereditary retinal degeneration owing to Nr2e3 mutation) with mice carrying the macrophage Fas-induced apoptosis (Mafia) transgene, we generated double-mutant mice and studied the role of the resident retinal microglia. Microglial cells in these double-mutant mice express enhanced green fluorescent protein (EGFP) and a suicide gene that can trigger Fas-mediated apoptosis via systemic treatment with AP20187 (FK506 dimerizer). We demonstrated that more than 80% of the EGFP+ cells in retinas from rd7/rd7;Tg/Tg mice express Iba-1 (a microglial marker), and resident microglia are still present in the retina because AP20187 does not cross the blood-brain barrier. Hence, only circulating bone marrow (BM)-derived microglia are depleted. Depletion of circulating BM-derived microglia accelerates retinal degeneration in rd7 mice. An increased number of autofluorescent (AF) spots is a consequence of resident microglia proliferation, which in turn establishes an inflammatory cytokine milieu via the upregulation of IL-1β, IL-6 and TNFα expression. This inflammation is likely to accelerate retinal degeneration. This study not only identifies inflammation as a crucial step in the pathogenesis of retinal degeneration, but also highlights the involvement of specific cytokine genes that could serve as future treatment targets in retinal degenerations.
Collapse
Affiliation(s)
- Nan-Kai Wang
- Department of Anatomy and Cell Biology, National Taiwan University, Taipei 100, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Clifford AB, Elnaggar AM, Robison RA, O'Neill K. Investigating the role of macrophages in tumor formation using a MaFIA mouse model. Oncol Rep 2013; 30:890-6. [PMID: 23722325 DOI: 10.3892/or.2013.2508] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 05/15/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor-associated macrophages (TAMs) interact with tumors in their development, growth and metastatic activities. Using a transgenic mouse model that allows for the selective depletion of macrophages we were able to access the macrophage's potential to facilitate metastasis. In the MaFIA (Macrophage Fas-Induced Apoptosis) mouse, transgene-expressing cells of the myeloid lineage undergo death by apoptosis in the presence of the drug AP20187. Enhanced green fluorescent protein (EGFP) was fused to the suicide gene to allow identification of transgene-expressing cells. Tumor induction was accomplished by subdermal and intravenous injections of B16-F10 melanoma cells. Metastasis in mice with depleted macrophages was compared to metastasis in normal control mice. The lungs and kidneys were examined for metastatic cells. The macrophage-depleted groups showed significantly less metastasis (P>0.001) compared to the control groups. We theorize that macrophages may aid the metastatic process by fusing with melanoma cells. Using appropriate cell markers and fluorescence-activated cell sorting, we were able to detect a small population of double-positive cells. We confirmed cell fusion by microscopic analysis, visualizing the cell's morphology by both immunohistochemistry and immunofluorescence. The presence of double-positive cells suggests macrophage/cancer cell fusion could be a possible mechanism for metastasis.
Collapse
Affiliation(s)
- A B Clifford
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | | | | | | |
Collapse
|
176
|
Oligodendrocyte-specific activation of PERK signaling protects mice against experimental autoimmune encephalomyelitis. J Neurosci 2013; 33:5980-91. [PMID: 23554479 DOI: 10.1523/jneurosci.1636-12.2013] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
There is compelling evidence that oligodendrocyte apoptosis, in response to CNS inflammation, contributes significantly to the development of the demyelinating disorder multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). Therefore, approaches designed to protect oligodendrocytes would likely have therapeutic value. Activation of pancreatic endoplasmic reticulum kinase (PERK) signaling in response to endoplasmic reticulum (ER) stress increases cell survival under various cytotoxic conditions. Moreover, there is evidence that PERK signaling is activated in oligodendrocytes within demyelinating lesions in multiple sclerosis and EAE. Our previous study demonstrated that CNS delivery of the inflammatory cytokine interferon-γ before EAE onset protected mice against EAE, and this protection was dependent on PERK signaling. In our current study, we sought to elucidate the role of PERK signaling in oligodendrocytes during EAE. We generated transgenic mice that allow for temporally controlled activation of PERK signaling, in the absence of ER stress, specifically in oligodendrocytes. We demonstrated that persistent activation of PERK signaling was not deleterious to oligodendrocyte viability or the myelin of adult animals. Importantly, we found that enhanced activation of PERK signaling specifically in oligodendrocytes significantly attenuated EAE disease severity, which was associated with reduced oligodendrocyte apoptosis, demyelination, and axonal degeneration. This effect was not the result of an altered degree of the inflammatory response in EAE mice. Our results provide direct evidence that activation of PERK signaling in oligodendrocytes is cytoprotective, protecting mice against EAE.
Collapse
|
177
|
Howland SW, Poh CM, Gun SY, Claser C, Malleret B, Shastri N, Ginhoux F, Grotenbreg GM, Rénia L. Brain microvessel cross-presentation is a hallmark of experimental cerebral malaria. EMBO Mol Med 2013; 5:984-99. [PMID: 23681698 PMCID: PMC3721469 DOI: 10.1002/emmm.201202273] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 01/02/2023] Open
Abstract
Cerebral malaria is a devastating complication of Plasmodium falciparum infection. Its pathogenesis is complex, involving both parasite- and immune-mediated events. CD8+ T cells play an effector role in murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection. We have identified a highly immunogenic CD8 epitope in glideosome-associated protein 50 that is conserved across rodent malaria species. Epitope-specific CD8+ T cells are induced during PbA infection, migrating to the brain just before neurological signs manifest. They are functional, cytotoxic and can damage the blood–brain barrier in vivo. Such CD8+ T cells are also found in the brain during infection with parasite strains/species that do not induce neuropathology. We demonstrate here that PbA infection causes brain microvessels to cross-present parasite antigen, while non-ECM-causing parasites do not. Further, treatment with fast-acting anti-malarial drugs before the onset of ECM reduces parasite load and thus antigen presentation in the brain, preventing ECM death. Thus our data suggest that combined therapies targeting both the parasite and host antigen-presenting cells may improve the outcome of CM patients.
Collapse
Affiliation(s)
- Shanshan W Howland
- Singapore Immunology Network, Agency for Science, Technology and Research A*STAR, Singapore, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Sphingosine-1-phosphate-mediated osteoclast precursor monocyte migration is a critical point of control in antibone-resorptive action of active vitamin D. Proc Natl Acad Sci U S A 2013; 110:7009-13. [PMID: 23569273 DOI: 10.1073/pnas.1218799110] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The migration and positioning of osteoclast precursor monocytes are controlled by the blood-enriched lipid mediator sphingosine-1-phosphate (S1P) and have recently been shown to be critical points of control in osteoclastogenesis and bone homeostasis. Here, we show that calcitriol, which is the hormonally active form of vitamin D, and its therapeutically used analog, eldecalcitol, inhibit bone resorption by modulating this mechanism. Vitamin D analogs have been used clinically for treating osteoporosis, although the mode of its pharmacologic action remains to be fully elucidated. In this study, we found that active vitamin D reduced the expression of S1PR2, a chemorepulsive receptor for blood S1P, on circulating osteoclast precursor monocytes both in vitro and in vivo. Calcitriol- or eldecalcitol-treated monocytoid RAW264.7 cells, which display osteoclast precursor-like properties, migrated readily to S1P. Concordantly, the mobility of circulating CX3CR1(+) osteoclast precursor monocytes was significantly increased on systemic administration of active vitamin D. These results show a mechanism for active vitamin D in controlling the migratory behavior of circulating osteoclast precursors, and this action should be conducive to limiting osteoclastic bone resorption in vivo.
Collapse
|
179
|
Konduri V, Decker WK, Halpert MM, Gilbert B, Safdar A. Modeling dendritic cell vaccination for influenza prophylaxis: potential applications for niche populations. J Infect Dis 2013; 207:1764-72. [PMID: 23460750 DOI: 10.1093/infdis/jit087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cancer patients can exhibit negligible responses to prophylactic vaccinations, including influenza vaccination. To help address this issue, we developed in vitro and in vivo models of dendritic cell (DC) immunotherapy for the prevention of influenza virus infection. METHODS Human cord blood (CB)-derived or mouse splenocyte-derived DCs were loaded with purified recombinant hemagglutinin (rHA). T-cell responses to HA-loaded CB-derived DCs were determined by ELISpot. Protective efficacy was determined by vaccination of BALB/c mice with a single injection of 10(6) autologous DCs. DC migration to peripheral lymphoid organs was verified by carboxyfluorescein succinimidyl ester staining, and HA-specific antibody titers were determined by enzyme-linked immunosorbent assay. Mice were then challenged intranasally with BALB/c-adapted A/New Caledonia influenza virus derived from four consecutive lung pool passages. Antigen-presenting cell (APC) dysfunction was modeled using the MAFIA transgenic system, in which the Csf1r promoter conditionally drives AP20178-inducible Fas. RESULTS CB-derived human DCs were able to generate de novo T-cell responses against rHA, as determined by a system of rigorous controls. Mice vaccinated intraperitoneally developed HA titers detectable at serum dilutions of >1:1000. HA seroconverters survived virus challenge, whereas unvaccinated controls and vaccinated nonseroconverters lost weight and died. Furthermore, use of a model of APC-specific immunosuppression revealed that DC vaccination could generate HA-specific antibody titers under conditions in which protein vaccination could not. CONCLUSIONS The model demonstrates that DC immunotherapy for the prevention of influenza is feasible, and studies are underway to determine whether populations of immunosuppressed individuals might ultimately benefit from the procedure.
Collapse
Affiliation(s)
- Vanaja Konduri
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
180
|
Li J, Hsu HC, Mountz JD. Managing macrophages in rheumatoid arthritis by reform or removal. Curr Rheumatol Rep 2013; 14:445-54. [PMID: 22855296 DOI: 10.1007/s11926-012-0272-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophages play a central role in the pathogenesis of rheumatoid arthritis (RA). There is an imbalance of inflammatory and antiinflammatory macrophages in RA synovium. Although the polarization and heterogeneity of macrophages in RA have not been fully uncovered, the identity of macrophages in RA can potentially be defined by their products, including the co-stimulatory molecules, scavenger receptors, different cytokines/chemokines and receptors, and transcription factors. In the last decade, efforts to understand the polarization, apoptosis regulation, and novel signaling pathways in macrophages, as well as how distinct activated macrophages influence disease progression, have led to strategies that target macrophages with varied specificity and selectivity. Major targets that are related to macrophage development and apoptosis include TNF-α, IL-1, IL-6, GM-CSF, M-CSF, death receptor 5 (DR5), Fas, and others, as listed in Table 1. Combined data from clinical, preclinical, and animal studies of inhibitors of these targets have provided valuable insights into their roles in the disease progression and, subsequently, have led to the evolving therapeutic paradigms in RA. In this review, we propose that reestablishment of macrophage equilibrium by inhibiting the development of, and/or eliminating, the proinflammatory macrophages will be an effective therapeutic approach for RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Jun Li
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, SHEL Bldg., Suite 337, 1825 University Blvd., Birmingham, AL 35294-2182, USA.
| | | | | |
Collapse
|
181
|
Luo J, Elwood F, Britschgi M, Villeda S, Zhang H, Ding Z, Zhu L, Alabsi H, Getachew R, Narasimhan R, Wabl R, Fainberg N, James ML, Wong G, Relton J, Gambhir SS, Pollard JW, Wyss-Coray T. Colony-stimulating factor 1 receptor (CSF1R) signaling in injured neurons facilitates protection and survival. ACTA ACUST UNITED AC 2013; 210:157-72. [PMID: 23296467 PMCID: PMC3549715 DOI: 10.1084/jem.20120412] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Colony-stimulating factor 1 and IL-34 protect against and partially reverse neurodegeneration in mice in part via promoting CREB signaling. Colony-stimulating factor 1 (CSF1) and interleukin-34 (IL-34) are functional ligands of the CSF1 receptor (CSF1R) and thus are key regulators of the monocyte/macrophage lineage. We discovered that systemic administration of human recombinant CSF1 ameliorates memory deficits in a transgenic mouse model of Alzheimer’s disease. CSF1 and IL-34 strongly reduced excitotoxin-induced neuronal cell loss and gliosis in wild-type mice when administered systemically before or up to 6 h after injury. These effects were accompanied by maintenance of cAMP responsive element–binding protein (CREB) signaling in neurons rather than in microglia. Using lineage-tracing experiments, we discovered that a small number of neurons in the hippocampus and cortex express CSF1R under physiological conditions and that kainic acid–induced excitotoxic injury results in a profound increase in neuronal receptor expression. Selective deletion of CSF1R in forebrain neurons in mice exacerbated excitotoxin-induced death and neurodegeneration. We conclude that CSF1 and IL-34 provide powerful neuroprotective and survival signals in brain injury and neurodegeneration involving CSF1R expression on neurons.
Collapse
Affiliation(s)
- Jian Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Abstract
Primary cultures are an important in vitro tool to study cellular processes and interactions. These cultures are complex systems, composed of many cell types, including neurons, astrocytes, oligodendrocytes, microglia, NG2 cells, and endothelial cells. For some studies it is necessary to be able to study a pure culture of one cell type, or eliminate a particular cell type, to better understand its function. There exist cell culture protocols for making pure astrocyte or microglia cultures. Here we present two protocols to produce cultures depleted for microglia: in the first case, from a mixed astrocyte-microglia culture and, in the second, for eliminating microglia from neuronal cultures.
Collapse
|
183
|
Uittenbogaard AM, Chelvarajan RL, Myers-Morales T, Gorman AA, Brickey WJ, Ye Z, Kaplan AM, Cohen DA, Ting JPY, Straley SC. Toward a molecular pathogenic pathway for Yersinia pestis YopM. Front Cell Infect Microbiol 2012; 2:155. [PMID: 23248776 PMCID: PMC3518861 DOI: 10.3389/fcimb.2012.00155] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/22/2012] [Indexed: 11/13/2022] Open
Abstract
YopM is one of the six "effector Yops" of the human-pathogenic Yersinia, but its mechanism has not been defined. After delivery to J774A.1 monocyte-like cells, YopM can rapidly bind and activate the serine/threonine kinases RSK1 and PRK2. However, in infected mice, effects of Y. pestis YopM have been seen only after 24-48 h post-infection (p.i.). To identify potential direct effects of YopM in-vivo we tested for effects of YopM at 1 h and 16-18 h p.i. in mice infected systemically with 10(6) bacteria. At 16 h p.i., there was a robust host response to both parent and ΔyopM-1 Y. pestis KIM5. Compared to cells from non-infected mice, CD11b(+) cells from spleens of infected mice produced more than 100-fold greater IFNγ. In the corresponding sera there were more than 100-fold greater amounts of IFNγ, G-CSF, and CXCL9, as well as more than 10-fold greater amounts of IL-6, CXCL10, and CXCL1. The only YopM-related differences were slightly lower CXCL10 and IL-6 in sera from mice infected 16 h with parent compared to ΔyopM-1 Y. pestis. Microarray analysis of the CD11b(+) cells did not identify consistent transcriptional differences of ≥4-fold at 18 h p.i. However, at 1 h p.i. mRNA for early growth response transcription factor 1 (Egr1) was decreased when YopM was present. Bone marrow-derived macrophages infected for 1 h also expressed lower Egr1 message when YopM was present. Infected J774A.1 cells showed greater expression of Egr1 at 1 h p.i. when YopM was present, but this pattern reversed at 3 h. At 6 h p.i., Cxcl10 mRNA was lower in parent-strain infected cells. We conclude that decreased Egr1 expression is a very early transcriptional effect of YopM and speculate that a pathway may exist from RSK1 through Egr1. These studies revealed novel early transcriptional effects of YopM but point to a time after 18 h of infection when critical transitional events lead to later major effects on cytokine gene transcription.
Collapse
Affiliation(s)
- Annette M Uittenbogaard
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky Lexington, KY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Affiliation(s)
- Susan C Straley
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
185
|
Gheryani N, Coffelt SB, Gartland A, Rumney RMH, Kiss-Toth E, Lewis CE, Tozer GM, Greaves DR, Dear TN, Miller G. Generation of a novel mouse model for the inducible depletion of macrophages in vivo. Genesis 2012; 51:41-9. [PMID: 22927121 DOI: 10.1002/dvg.22343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 08/22/2012] [Accepted: 08/22/2012] [Indexed: 11/08/2022]
Abstract
Macrophages play an essential role in tissue homeostasis, innate immunity, inflammation, and wound repair. Macrophages are also essential during development, severely limiting the use of mouse models in which these cells have been constitutively deleted. Consequently, we have developed a transgenic model of inducible macrophage depletion in which macrophage-specific induction of the cytotoxic diphtheria toxin A chain (DTA) is achieved by administration of doxycycline. Induction of the DTA protein in transgenic animals resulted in a significant 50% reduction in CD68+ macrophages of the liver, spleen, and bone over a period of 6 weeks. Pertinently, the macrophages remaining after doxycycline treatment were substantially smaller and are functionally impaired as shown by reduced inflammatory cytokine production in response to lipopolysaccharide. This inducible model of macrophage depletion can now be utilized to determine the role of macrophages in both development and animal models of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Nabeia Gheryani
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Dhaliwal K, Scholefield E, Ferenbach D, Gibbons M, Duffin R, Dorward DA, Morris AC, Humphries D, MacKinnon A, Wilkinson TS, Wallace WAH, van Rooijen N, Mack M, Rossi AG, Davidson DJ, Hirani N, Hughes J, Haslett C, Simpson AJ. Monocytes control second-phase neutrophil emigration in established lipopolysaccharide-induced murine lung injury. Am J Respir Crit Care Med 2012; 186:514-24. [PMID: 22822022 DOI: 10.1164/rccm.201112-2132oc] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Acute lung injury (ALI) is an important cause of morbidity and mortality, with no currently effective pharmacological therapies. Neutrophils have been specifically implicated in the pathogenesis of ALI, and there has been significant research into the mechanisms of early neutrophil recruitment, but those controlling the later phases of neutrophil emigration that characterize disease are poorly understood. OBJECTIVES To determine the influence of peripheral blood monocytes (PBMs) in established ALI. METHODS In a murine model of LPS-induced ALI, three separate models of conditional monocyte ablation were used: systemic liposomal clodronate (sLC), inducible depletion using CD11b diphtheria toxin receptor (CD11b DTR) transgenic mice, and antibody-dependent ablation of CCR2(hi) monocytes. MEASUREMENTS AND MAIN RESULTS PBMs play a critical role in regulating neutrophil emigration in established murine LPS-induced lung injury. Gr1(hi) and Gr1(lo) PBM subpopulations contribute to this process. PBM depletion is associated with a significant reduction in measures of lung injury. The specificity of PBM depletion was demonstrated by replenishment studies in which the effects were reversed by systemic PBM infusion but not by systemic or local pulmonary infusion of mature macrophages or lymphocytes. CONCLUSIONS These results suggest that PBMs, or the mechanisms by which they influence pulmonary neutrophil emigration, could represent therapeutic targets in established ALI.
Collapse
Affiliation(s)
- Kevin Dhaliwal
- MRC Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Belnoue E, Tougne C, Rochat AF, Lambert PH, Pinschewer DD, Siegrist CA. Homing and adhesion patterns determine the cellular composition of the bone marrow plasma cell niche. THE JOURNAL OF IMMUNOLOGY 2012; 188:1283-91. [PMID: 22262758 DOI: 10.4049/jimmunol.1103169] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
According to commonly held concepts, plasma cell (PC) longevity in bone marrow (BM) depends upon their access to survival niches. These are thought to exist in nursery cell types, which support PCs by secreting PC survival factors. To better define PC survival niches and their functioning, we adoptively transferred traceable Blimp-1-(GFP) PCs into recipient mice lacking a proliferation-inducing ligand (APRIL), IL-6, or macrophage migration inhibitory factor. Transferred BMPCs were preferentially associated with Ly-6C(high) monocytes (normalized colocalization index: 9.84), eosinophils (4.29), and megakaryocytes (2.12). Although APRIL was essential for BMPC survival, PC recruitment into the proximity of nursery cells was unimpaired in APRIL-deficient mice, questioning the concept that the same factors account for attraction/retention of PCs as for their local survival. Rather, the order of colocalization with BMPCs (monocytes > eosinophils > megakaryocytes) reflected these cells' relative expression of CXCR4, VLA-4, and LFA-1, the homing and adhesion molecules that direct/retain PCs in the BM. This suggests a scenario wherein the cellular composition of the BMPC niche is defined by a common pattern of attraction/retention on CXCL12-abundant reticular docking cells. Thereby, PCs are directed to associate in a functional BM niche with hematopoietic CXCR4(+)VLA-4(+)LFA-1(+) nursery cells, which provide PC survival factors.
Collapse
Affiliation(s)
- Elodie Belnoue
- Department of Pathology-Immunology, World Health Organization Collaborating Center for Vaccinology and Neonatal Immunology, University of Geneva, CH-1211 Geneva 4, Switzerland.
| | | | | | | | | | | |
Collapse
|
188
|
Linch SN, Danielson ET, Kelly AM, Tamakawa RA, Lee JJ, Gold JA. Interleukin 5 is protective during sepsis in an eosinophil-independent manner. Am J Respir Crit Care Med 2012; 186:246-54. [PMID: 22652030 DOI: 10.1164/rccm.201201-0134oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
RATIONALE The immune response in sepsis is characterized by overt immune dysfunction. Studies indicate immunostimulation represents a viable therapy for patients. One study suggests a potentially protective role for interleukin 5 (IL-5) in sepsis; however, the loss of eosinophils in this disease presents a paradox. OBJECTIVES To assess the protective and eosinophil-independent effects of IL-5 in sepsis. METHODS We assessed the effects of IL-5 administration on survival, bacterial burden, and cytokine production after polymicrobial sepsis. In addition, we examined the effects on macrophage phagocytosis and survival using fluorescence microscopy and flow cytometry. MEASUREMENTS AND MAIN RESULTS Loss of IL-5 increased mortality and tissue damage in the lung, IL-6 and IL-10 production, and bacterial burden during sepsis. Therapeutic administration of IL-5 improved mortality in sepsis. Interestingly, IL-5 administration resulted in neutrophil recruitment in vivo. IL-5 overexpression in the absence of eosinophils resulted in decreased mortality from sepsis and increased circulating neutrophils and monocytes, suggesting their importance in the protective effects of IL-5. Furthermore, novel data demonstrate IL-5 receptor expression on neutrophils and monocytes in sepsis. IL-5 augmented cytokine secretion, activation, phagocytosis, and survival of macrophages. Importantly, macrophage depletion before the onset of sepsis eliminated IL-5-mediated protection. The protective effects of IL-5 were confirmed in humans, where IL-5 levels were elevated in patients with sepsis. Moreover, neutrophils and monocytes from patients expressed the IL-5 receptor. CONCLUSIONS Taken together, these data support a novel role for IL-5 on noneosinophilic myeloid populations, and suggest treatment with IL-5 may be a viable therapy for sepsis.
Collapse
Affiliation(s)
- Stefanie N Linch
- Department of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon 97216, USA
| | | | | | | | | | | |
Collapse
|
189
|
Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, Leboeuf M, Low D, Oller G, Almeida F, Choy SHY, Grisotto M, Renia L, Conway SJ, Stanley ER, Chan JKY, Ng LG, Samokhvalov IM, Merad M, Ginhoux F. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. ACTA ACUST UNITED AC 2012; 209:1167-81. [PMID: 22565823 PMCID: PMC3371735 DOI: 10.1084/jem.20120340] [Citation(s) in RCA: 578] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Langerhans cells (LCs) are the dendritic cells (DCs) of the epidermis, forming one of the first hematopoietic lines of defense against skin pathogens. In contrast to other DCs, LCs arise from hematopoietic precursors that seed the skin before birth. However, the origin of these embryonic precursors remains unclear. Using in vivo lineage tracing, we identify a first wave of yolk sac (YS)-derived primitive myeloid progenitors that seed the skin before the onset of fetal liver hematopoiesis. YS progenitors migrate to the embryo proper, including the prospective skin, where they give rise to LC precursors, and the brain rudiment, where they give rise to microglial cells. However, in contrast to microglia, which remain of YS origin throughout life, YS-derived LC precursors are largely replaced by fetal liver monocytes during late embryogenesis. Consequently, adult LCs derive predominantly from fetal liver monocyte-derived cells with a minor contribution of YS-derived cells. Altogether, we establish that adult LCs have a dual origin, bridging early embryonic and late fetal myeloid development.
Collapse
Affiliation(s)
- Guillaume Hoeffel
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Immunos Building #3-4, BIOPOLIS, 138648, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Nanoinjection: pronuclear DNA delivery using a charged lance. Transgenic Res 2012; 21:1279-90. [PMID: 22415347 DOI: 10.1007/s11248-012-9610-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/02/2012] [Indexed: 02/06/2023]
Abstract
We present a non-fluidic pronuclear injection method using a silicon microchip "nanoinjector" composed of a microelectromechanical system with a solid, electrically conductive lance. Unlike microinjection which uses fluid delivery of DNA, nanoinjection electrically accumulates DNA on the lance, the DNA-coated lance is inserted into the pronucleus, and DNA is electrically released. We compared nanoinjection and microinjection side-by-side over the course of 4 days, injecting 1,013 eggs between the two groups. Nanoinjected zygotes had significantly higher rates of integration per injected embryo, with 6.2% integration for nanoinjected embryos compared to 1.6% integration for microinjected embryos. This advantage is explained by nanoinjected zygotes' significantly higher viability in two stages of development: zygote progress to two-cell stage, and progress from two-cell stage embryos to birth. We observed that 77.6% of nanoinjected zygotes proceeded to two-cell stage compared to 54.7% of microinjected zygotes. Of the healthy two-cell stage embryos, 52.4% from the nanoinjection group and 23.9% from the microinjected group developed into pups. Structural advantages of the nanoinjector are likely to contribute to the high viability observed. For instance, because charge is used to retain and release DNA, extracellular fluid is not injected into the pronucleus and the cross-sectional area of the nanoinjection lance (0.06 µm(2)) is smaller than that of a microinjection pipette tip (0.78 µm(2)). According to results from the comparative nanoinjection versus microinjection study, we conclude that nanoinjection is a viable method of pronuclear DNA transfer which presents viability advantages over microinjection.
Collapse
|
191
|
Sun Y, Karmakar M, Taylor PR, Rietsch A, Pearlman E. ExoS and ExoT ADP ribosyltransferase activities mediate Pseudomonas aeruginosa keratitis by promoting neutrophil apoptosis and bacterial survival. THE JOURNAL OF IMMUNOLOGY 2012; 188:1884-95. [PMID: 22250085 DOI: 10.4049/jimmunol.1102148] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pseudomonas aeruginosa is a leading cause of blinding corneal ulcers worldwide. To determine the role of type III secretion in the pathogenesis of P. aeruginosa keratitis, corneas of C57BL/6 mice were infected with P. aeruginosa strain PAO1 or PAK, which expresses ExoS, ExoT, and ExoY, but not ExoU. PAO1- and PAK-infected corneas developed severe disease with pronounced opacification and rapid bacterial growth. In contrast, corneas infected with ΔpscD or ΔpscJ mutants that cannot assemble a type III secretion system, or with mutants lacking the translocator proteins, do not develop clinical disease, and bacteria are rapidly killed by infiltrating neutrophils. Furthermore, survival of PAO1 and PAK strains in the cornea and development of corneal disease was impaired in ΔexoS, ΔexoT, and ΔexoST mutants of both strains, but not in a ΔexoY mutant. ΔexoST mutants were also rapidly killed in neutrophils in vitro and were impaired in their ability to promote neutrophil apoptosis in vivo compared with PAO1. Point mutations in the ADP ribosyltransferase (ADPR) regions of ExoS or ExoT also impaired proapoptotic activity in infected neutrophils, and exoST(ADPR-) mutants replicated the ΔexoST phenotype in vitro and in vivo, whereas mutations in rho-GTPase-activating protein showed the same phenotype as PAO1. Together, these findings demonstrate that the pathogenesis of P. aeruginosa keratitis in ExoS- and ExoT-producing strains is almost entirely due to their ADPR activities, which subvert the host response by targeting the antibacterial activity of infiltrating neutrophils.
Collapse
Affiliation(s)
- Yan Sun
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
192
|
O'Brien J, Martinson H, Durand-Rougely C, Schedin P. Macrophages are crucial for epithelial cell death and adipocyte repopulation during mammary gland involution. Development 2012; 139:269-75. [DOI: 10.1242/dev.071696] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mammary gland development is dependent on macrophages, as demonstrated by their requirement during the expansion phases of puberty and pregnancy. Equally dramatic tissue restructuring occurs following lactation, when the gland regresses to a state that histologically resembles pre-pregnancy through massive programmed epithelial cell death and stromal repopulation. Postpartum involution is characterized by wound healing-like events, including an influx of macrophages with M2 characteristics. Macrophage levels peak after the initial wave of epithelial cell death, suggesting that initiation and execution of cell death are macrophage independent. To address the role of macrophages during weaning-induced mammary gland involution, conditional systemic deletion of macrophages expressing colony stimulating factor 1 receptor (CSF1R) was initiated just prior to weaning in the Mafia mouse model. Depletion of CSF1R+ macrophages resulted in delayed mammary involution as evidenced by loss of lysosomal-mediated and apoptotic epithelial cell death, lack of alveolar regression and absence of adipocyte repopulation 7 days post-weaning. Failure to execute involution occurred in the presence of milk stasis and STAT3 activation, indicating that neither is sufficient to initiate involution in the absence of CSF1R+ macrophages. Injection of wild-type bone marrow-derived macrophages (BMDMs) or M2-differentiated macrophages into macrophage-depleted mammary glands was sufficient to rescue involution, including apoptosis, alveolar regression and adipocyte repopulation. BMDMs exposed to the postpartum mammary involution environment upregulated the M2 markers arginase 1 and mannose receptor. These data demonstrate the necessity of macrophages, and implicate M2-polarized macrophages, for epithelial cell death during normal postpartum mammary gland involution.
Collapse
Affiliation(s)
- Jenean O'Brien
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave, Aurora, CO 80045, USA
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, MS8104, RC-1S, 5117, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Holly Martinson
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave, Aurora, CO 80045, USA
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, MS8104, RC-1S, 5117, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Clarissa Durand-Rougely
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave, Aurora, CO 80045, USA
| | - Pepper Schedin
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave, Aurora, CO 80045, USA
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, MS8104, RC-1S, 5117, 12801 E 17th Ave, Aurora, CO 80045, USA
- University of Colorado Cancer Center, Bldg 500, Suite 6004C, 13001 E 17th Place, Aurora, CO 80045 USA
- AMC Cancer Research Center, Bldg 500, Suite 6004C, 13001 E 17th Place, Aurora, CO 80045, USA
| |
Collapse
|
193
|
Ishii T, Kawamura S, Nishiyama I, Kikuta J, Ishii M. Use of intravital microscopy and in vitro chemotaxis assays to study the roles of sphingosine-1-phosphate in bone homeostasis. Methods Mol Biol 2012; 874:129-139. [PMID: 22528444 DOI: 10.1007/978-1-61779-800-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
We describe a method to visualize the migration of osteoclast precursors within intact murine bone -marrow in real time using intravital multiphoton microscopy. Conventionally, cell migration has been evaluated using in vitro systems, such as transmigration assays. Although these methods are convenient for quantification and are highly reproducible, these in vitro assay systems may not accurately reflect in vivo cellular behavior. In addition to in vitro analyses, recent technological progress in two-photon excitation-based laser microscopy has enabled the visualization of dynamic cell behavior deep inside intact living organs. Combining this imaging method with in vitro chemoattraction analyses, we have revealed that sphingosine-1-phosphate (S1P), a lipid mediator enriched in blood, bidirectionally controls the trafficking of osteoclast precursors between the circulation and bone marrow cavities via G protein-coupled receptors (GPCRs).
Collapse
Affiliation(s)
- Taeko Ishii
- Laboratory of Cellular Dynamics, WPI-Immunology Frontier Research Center, Osaka University, Suita, Japan
| | | | | | | | | |
Collapse
|
194
|
Mountz JD, Li J, Hsu HC. Systemic autoimmunity caused by fas deficiency in macrophages: a new perspective on the first identified autoimmunity gene. ACTA ACUST UNITED AC 2011; 64:609-12. [PMID: 22139895 DOI: 10.1002/art.34321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
195
|
Chinnery HR, McLenachan S, Binz N, Sun Y, Forrester JV, Degli-Esposti MA, Pearlman E, McMenamin PG. TLR9 ligand CpG-ODN applied to the injured mouse cornea elicits retinal inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:209-20. [PMID: 22085974 DOI: 10.1016/j.ajpath.2011.09.041] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 09/11/2011] [Accepted: 09/14/2011] [Indexed: 11/15/2022]
Abstract
During bacterial and viral infections, unmethylated CpG-DNA released by proliferating and dying microbes is recognized by toll-like receptor (TLR) 9 in host cells, initiating innate immune responses. Many corneal infections occur secondary to epithelial breaches and represent a major cause of vision impairment and blindness globally. To mimic this clinical situation, we investigated mechanisms of TLR9 ligand-induced corneal inflammation in mice after epithelial debridement. Application of CpG oligodeoxynucleotides (ODNs) resulted in neutrophil and macrophage infiltration to the cornea and loss of transparency. By 6 hours after CpG-ODN administration, TLR9 mRNA was increased in the cornea and retina. In vivo clinical examination at 24 hours revealed inflammatory infiltrates in the vitreous and retina, which were confirmed ex vivo to be neutrophils and macrophages, along with activated resident microglia. CpG-ODN-induced intraocular inflammation was abrogated in TLR9(-/-) and macrophage-depleted mice. Bone marrow reconstitution of irradiated TLR9(-/-) mice with TLR9(+/+) bone marrow led to restored corneal inflammatory responses to CpG-ODN. Fluorescein isothiocyanate-CpG-ODN rapidly penetrated the cornea and ocular media to reach the retina, where it was present within CD68(+) retinal macrophages and microglia. These data show that topically applied CpG-ODN induces intraocular inflammation owing to TLR9 activation of monocyte-lineage cells. These novel findings indicate that microbial CpG-DNA released during bacterial and/or viral keratitis can cause widespread inflammation within the eye, including the retina.
Collapse
Affiliation(s)
- Holly R Chinnery
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
196
|
Fischer MA, Davies ML, Reider IE, Heipertz EL, Epler MR, Sei JJ, Ingersoll MA, Van Rooijen N, Randolph GJ, Norbury CC. CD11b⁺, Ly6G⁺ cells produce type I interferon and exhibit tissue protective properties following peripheral virus infection. PLoS Pathog 2011; 7:e1002374. [PMID: 22102816 PMCID: PMC3213107 DOI: 10.1371/journal.ppat.1002374] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 09/28/2011] [Indexed: 12/22/2022] Open
Abstract
The goal of the innate immune system is containment of a pathogen at the site of infection prior to the initiation of an effective adaptive immune response. However, effector mechanisms must be kept in check to combat the pathogen while simultaneously limiting undesirable destruction of tissue resulting from these actions. Here we demonstrate that innate immune effector cells contain a peripheral poxvirus infection, preventing systemic spread of the virus. These innate immune effector cells are comprised primarily of CD11b+Ly6C+Ly6G- monocytes that accumulate initially at the site of infection, and are then supplemented and eventually replaced by CD11b+Ly6C+Ly6G+ cells. The phenotype of the CD11b+Ly6C+Ly6G+ cells resembles neutrophils, but the infiltration of neutrophils typically occurs prior to, rather than following, accumulation of monocytes. Indeed, it appears that the CD11b+Ly6C+Ly6G+ cells that infiltrated the site of VACV infection in the ear are phenotypically distinct from the classical description of both neutrophils and monocyte/macrophages. We found that CD11b+Ly6C+Ly6G+ cells produce Type I interferons and large quantities of reactive oxygen species. We also observed that depletion of Ly6G+ cells results in a dramatic increase in tissue damage at the site of infection. Tissue damage is also increased in the absence of reactive oxygen species, although reactive oxygen species are typically thought to be damaging to tissue rather than protective. These data indicate the existence of a specialized population of CD11b+Ly6C+Ly6G+ cells that infiltrates a site of virus infection late and protects the infected tissue from immune-mediated damage via production of reactive oxygen species. Regulation of the action of this population of cells may provide an intervention to prevent innate immune-mediated tissue destruction. During a natural virus infection, small doses of infectious virus are deposited at a peripheral infection site, and then a “race” ensues, in which the replicating virus attempts to “outpace” the responding immune system of the host. In the early phases of infection, the innate immune system must contain the infection prior to the development of an effective adaptive response. Here we have characterized the cells of the innate immune system that move to a site of peripheral virus infection, and we find that a subset of these cells display atypical expression of cell surface molecules, timing of infiltration, and function. These cells protect the infected tissue from damage by producing reactive oxygen molecules, which are widely accepted to increase tissue damage. Therefore our findings indicate that during a peripheral virus infection, the typical rules governing the function of the innate immune system are altered to prevent tissue damage.
Collapse
Affiliation(s)
- Matthew A. Fischer
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Michael L. Davies
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Irene E. Reider
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Erica L. Heipertz
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Melanie R. Epler
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Janet J. Sei
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Molly A. Ingersoll
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Nico Van Rooijen
- Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, Amsterdam, The Netherlands
| | - Gwendalyn J. Randolph
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Christopher C. Norbury
- Department of Microbiology and Immunology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
197
|
Abstract
The mononuclear phagocyte system (MPS) comprises monocytes, macrophages and dendritic cells. Tissue phagocytes share several cell surface markers, phagocytic capability and myeloid classification; however, the factors that regulate the differentiation, homeostasis and function of macrophages and dendritic cells remain largely unknown. The purpose of this manuscript is to review the tools that are currently available and those that are under development to study the origin and function of mononuclear phagocytes.
Collapse
|
198
|
Aldrich A, Kielian T. Central nervous system fibrosis is associated with fibrocyte-like infiltrates. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2952-62. [PMID: 22015460 DOI: 10.1016/j.ajpath.2011.08.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 08/12/2011] [Accepted: 08/18/2011] [Indexed: 12/25/2022]
Abstract
Fibrotic wall formation is essential for limiting pathogen dissemination during brain abscess development. However, little is known about the regulation of fibrotic processes in the central nervous system (CNS). Most CNS injury responses are associated with hypertrophy of resident astrocytes, a process termed reactive gliosis. Studies of fibrosis outside the CNS have identified two bone marrow-derived cell types, fibrocytes and alternatively activated M2 macrophages, as key mediators of fibrosis. The current study used bone marrow chimeras generated from green fluorescent protein transgenic mice to evaluate the appearance of these cell types and whether bone marrow-derived cells were capable of acquiring fibrotic characteristics during brain abscess development. Immunofluorescence staining revealed partial overlap between green fluorescent protein, α-smooth muscle actin, and procollagen, suggesting that a population of cells forming the brain abscess capsule originate from a bone marrow precursor. In addition, the influx of fibrocyte-like cells into brain abscesses immediately preceded the onset of fibrotic encapsulation. Fibrotic wall formation was also associated with increased numbers of alternatively activated M2 microglia and macrophages. To our knowledge, this is the first study demonstrating that bone marrow-derived infiltrates are capable of expressing fibrotic molecules during CNS inflammation.
Collapse
Affiliation(s)
- Amy Aldrich
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5900, USA
| | | |
Collapse
|
199
|
Hart KM, Byrne KT, Molloy MJ, Usherwood EM, Berwin B. IL-10 immunomodulation of myeloid cells regulates a murine model of ovarian cancer. Front Immunol 2011; 2:29. [PMID: 22566819 PMCID: PMC3342001 DOI: 10.3389/fimmu.2011.00029] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 07/07/2011] [Indexed: 12/31/2022] Open
Abstract
Elevated levels of IL-10 in the microenvironment of human ovarian cancer and murine models of ovarian cancer are well established and correlate with poor clinical prognosis. However, amongst a myriad of immunosuppressive factors, the actual contribution of IL-10 to the ovarian tumor microenvironment, the mechanisms by which it acts, and its possible functional redundancy are unknown. We previously demonstrated that elimination of the myeloid-derived suppressor cell (MDSC) compartment within the ovarian tumor ascites inhibited tumor progression and, intriguingly, significantly decreased local IL-10 levels. Here we identify a novel pathway in which the tumor-infiltrating MDSC are the predominant producers of IL-10 and, importantly, require it to develop their immunosuppressive function in vivo. Importantly, we demonstrate that the role of IL-10 is critical, and not redundant with other immunosuppressive molecules, to in vivo tumor progression: blockade of the IL-10 signaling network results in alleviation of MDSC-mediated immunosuppression, altered T cell phenotype and activity, and improved survival. These studies define IL-10 as a fundamental modulator of both MDSC and T cells within the ovarian tumor microenvironment. Importantly, IL-10 signaling is shown to be necessary to the development and maintenance of a permissive tumor microenvironment and represents a viable target for anti-tumor strategies.
Collapse
Affiliation(s)
- Kevin M Hart
- Berwin Laboratory, Department of Microbiology and Immunology, Dartmouth Medical Center Lebanon, NH, USA
| | | | | | | | | |
Collapse
|
200
|
Ovarian tumor-induced T cell suppression is alleviated by vascular leukocyte depletion. Transl Oncol 2011; 2:291-9. [PMID: 19956391 DOI: 10.1593/tlo.09190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 07/16/2009] [Accepted: 07/27/2009] [Indexed: 11/18/2022] Open
Abstract
The ovarian cancer microenvironment recruits an array of immune cells to the site of tumor growth. Within the peritoneal ascites of both humans and mice, the predominant population of tumor-infiltrating leukocytes is a CD11c(+)CD11b(+) population variably referred to as vascular leukocytes (VLCs), tumor-associated macrophages, and immature dendritic cells. We have previously shown that these cells are critical for tumor growth because their selective elimination from the peritoneal tumor microenvironment inhibited tumor progression. However, the underlying mechanism by which this therapy was efficacious is poorly understood. Here, we use the murine ID8 ovarian tumor model to demonstrate that the tumor microenvironment induces in vivo immunosuppression of T cells and that the SR-A(+) VLCs mediate this suppression. Importantly, the elimination of SR-A(+) VLCs from the peritoneum of tumor-bearing mice relieves the T cell suppression. Moreover, the profound changes that VLC elimination has on the immune system are T cell-dependent because the protective antitumor effect of VLC elimination does not occur when CD8 T cells are concomitantly depleted. These results were confirmed and extended with the use of a genetic model for VLC depletion, which demonstrated that short-term therapeutic depletion of VLCs alleviates immunosuppression and allows for efficacious vaccination against model tumor antigens in tumor-bearing mice. These studies provide a mechanistic explanation for how leukocytes contribute to ovarian tumor progression and, correspondingly, how leukocyte depletion inhibits tumor growth.
Collapse
|