101
|
Gil-Pulido J, Zernecke A. Antigen-presenting dendritic cells in atherosclerosis. Eur J Pharmacol 2017; 816:25-31. [DOI: 10.1016/j.ejphar.2017.08.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 11/29/2022]
|
102
|
Subramanian N, Wu Z, Reister F, Sampaio KL, Frascaroli G, Cicin-Sain L, Mertens T. Naïve T cells are activated by autologous HCMV-infected endothelial cells through NKG2D and can control HCMV transmission in vitro. J Gen Virol 2017; 98:3068-3085. [PMID: 29165229 DOI: 10.1099/jgv.0.000976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Apart from classical antigen-presenting cells (APCs) like dendritic cells and macrophages, there are semiprofessional APCs such as endothelial cells (ECs) and Langerhans' cells. Human cytomegalovirus (HCMV) infects a wide range of cell types including the ECs which are involved in the trafficking and homing of T cells. By investigating the interaction of naïve T cells obtained from HCMV-seronegative umbilical cord blood with autologous HCMV-infected human umbilical vein ECs (HUVECs), we could show that the activation of naïve T cells occurred after 1 day of peripheral blood mononuclear cell (PBMC) exposure to HCMV-infected HUVECs. The percentage of activated T cells increased over time and the activation of naïve T cells was not induced by either autologous uninfected HUVECs or by autologous HCMV-infected fibroblasts. The activation of T cells occurred also when purified T cells were co-cultured with HCMV-infected HUVECs. In addition, in most of the donors only CD8+ T cells were activated, when the purified T cells were exposed to HCMV-infected HUVECs. The activation of naïve T cells was inhibited when the NKG2D receptor was blocked on the surface of T cells and among the different NKG2D ligands, we identified two ligands (ULBP4 and MICA) on HCMV-infected HUVECs which might be the interaction partners of the NKG2D receptor. Using a functional cell culture assay, we could show that these activated naïve T cells specifically inhibited HCMV transmission. Altogether, we identified a novel specific activation mechanism of naïve T cells from the umbilical cord by HCMV-infected autologous HUVECs through interaction with NKG2D.
Collapse
Affiliation(s)
| | - Zeguang Wu
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Frank Reister
- Gynecology and Obstetrics Clinics, Ulm University Hospital, Ulm, Germany
| | | | - Giada Frascaroli
- Institute of Virology, Ulm University Medical Center, Ulm, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Location Hannover-Braunschweig, Germany
| | - Thomas Mertens
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
103
|
Fu X, He F, Li Y, Shahveranov A, Hutchins AP. Genomic and molecular control of cell type and cell type conversions. CELL REGENERATION 2017; 6:1-7. [PMID: 29348912 PMCID: PMC5769489 DOI: 10.1016/j.cr.2017.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/06/2017] [Accepted: 09/18/2017] [Indexed: 12/17/2022]
Abstract
Organisms are made of a limited number of cell types that combine to form higher order tissues and organs. Cell types have traditionally been defined by their morphologies or biological activity, yet the underlying molecular controls of cell type remain unclear. The onset of single cell technologies, and more recently genomics (particularly single cell genomics), has substantially increased the understanding of the concept of cell type, but has also increased the complexity of this understanding. These new technologies have added a new genome wide molecular dimension to the description of cell type, with genome-wide expression and epigenetic data acting as a cell type ‘fingerprint’ to describe the cell state. Using these genomic fingerprints cell types are being increasingly defined based on specific genomic and molecular criteria, without necessarily a distinct biological function. In this review, we will discuss the molecular definitions of cell types and cell type control, and particularly how endogenous and exogenous transcription factors can control cell types and cell type conversions.
Collapse
|
104
|
Ueffing K, Abberger H, Westendorf AM, Matuschewski K, Buer J, Hansen W. Conventional CD11c high Dendritic Cells Are Important for T Cell Priming during the Initial Phase of Plasmodium yoelii Infection, but Are Dispensable at Later Time Points. Front Immunol 2017; 8:1333. [PMID: 29085373 PMCID: PMC5650681 DOI: 10.3389/fimmu.2017.01333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are highly specialized antigen-presenting cells that orchestrate adaptive immune responses to pathogens. During malaria infection pro- and anti-inflammatory T cell responses have to be tightly balanced to ensure parasite clearance without induction of severe immune pathologies. However, the precise role of CD11chigh DCs in this process is still discussed controversially. Here, we demonstrate that long-term depletion of conventional CD11chigh DCs in Plasmodium yoelii (P. yoelii)-infected diphtheria toxin (DT)-treated RosaiDTR/CD11c-cre mice interferes with the activation of CD8+ and CD4+ T cells as well as CD4+Foxp3+ regulatory T cells at early time points during infection. Moreover, systemic levels of the pro-inflammatory cytokines IFN-γ and TNF-α were decreased in P. yoelii-infected mice deficient for CD11chigh DCs compared to infected RosaiDTR controls. To further elucidate the importance of CD11chigh DCs during the later phase of infection, we treated RosaiDTR/CD11c-cre and control mice with DT only from day 4 of P. yoelii infection onward. Strikingly, this approach had no impact on the activation and IFN-γ production of CD4+ and CD8+ effector T cells. These results indicate that CD11chigh DCs play a crucial role in eliciting effector T cell responses during the initial phase, but are dispensable during ongoing infection with P. yoelii.
Collapse
Affiliation(s)
- Kristina Ueffing
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Hanna Abberger
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kai Matuschewski
- Molecular Parasitology, Institute of Biology, Humboldt University of Berlin, Berlin, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
105
|
Désy O, Béland S, Vallin P, Riopel J, Latulippe E, Najafian N, Chandraker A, Agharazii M, Batal I, De Serres SA. IL-6 production by monocytes is associated with graft function decline in patients with borderline changes suspicious for acute T-cell-mediated rejection: a pilot study. Transpl Int 2017; 31:92-101. [DOI: 10.1111/tri.13070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/28/2017] [Accepted: 09/06/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Olivier Désy
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Stéphanie Béland
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Patrice Vallin
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Julie Riopel
- Department of Pathology; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Eva Latulippe
- Department of Pathology; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Nader Najafian
- Renal Division; Schuster Family Transplantation Research Center; Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School; Boston MA USA
| | - Anil Chandraker
- Renal Division; Schuster Family Transplantation Research Center; Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School; Boston MA USA
| | - Mohsen Agharazii
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| | - Ibrahim Batal
- Department of Pathology; Brigham and Women's Hospital and Harvard Medical School; Boston MA USA
| | - Sacha A. De Serres
- Transplantation Unit; Renal Division; Department of Medicine; University Health Center of Quebec; Faculty of Medicine; Laval University; Quebec QC Canada
| |
Collapse
|
106
|
Abstract
Dendritic cells (DCs) play critical roles in activating innate immune cells and initiating adaptive immune responses. The functions of DCs were originally obscured by their overlap with other mononuclear phagocytes, but new mouse models have allowed for the selective ablation of subsets of DCs and have helped to identify their non-redundant roles in the immune system. These tools have elucidated the functions of DCs in host defense against pathogens, autoimmunity, and cancer. This review will describe the mouse models generated to interrogate the role of DCs and will discuss how their use has progressively clarified our understanding of the unique functions of DC subsets.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
107
|
Belfield LA, Bennett JH, Abate W, Jackson SK. Exposure to Porphyromonas gingivalis LPS during macrophage polarisation leads to diminished inflammatory cytokine production. Arch Oral Biol 2017; 81:41-47. [DOI: 10.1016/j.archoralbio.2017.04.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/31/2017] [Accepted: 04/17/2017] [Indexed: 10/19/2022]
|
108
|
Abstract
The maintenance of monocytes, macrophages, and dendritic cells (DCs) involves manifold pathways of ontogeny and homeostasis that have been the subject of intense study in recent years. The concept of a peripheral mononuclear phagocyte system continually renewed by blood-borne monocytes has been modified to include specialized DC pathways of development that do not involve monocytes, and longevity through self-renewal of tissue macrophages. The study of development remains difficult owing to the plasticity of phenotypes and misconceptions about the fundamental structure of hematopoiesis. However, greater clarity has been achieved in distinguishing inflammatory monocyte-derived DCs from DCs arising in the steady state, and new concepts of conjoined lymphomyeloid hematopoiesis more easily accommodate the shared lymphoid and myeloid phenotypes of some DCs. Cross-species comparisons have also yielded coherent systems of nomenclature for all mammalian monocytes, macrophages, and DCs. Finally, the clear relationships between ontogeny and functional specialization offer information about the regulation of immune responses and provide new tools for the therapeutic manipulation of myeloid mononuclear cells in medicine.
Collapse
|
109
|
Porphyromonas gulae Activates Unprimed and Gamma Interferon-Primed Macrophages via the Pattern Recognition Receptors Toll-Like Receptor 2 (TLR2), TLR4, and NOD2. Infect Immun 2017. [PMID: 28630066 DOI: 10.1128/iai.00282-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gulae is an anaerobic, Gram-negative coccobacillus that has been associated with periodontal disease in companion animals. The aims of this study were to analyze the ligation of pattern recognition receptors by P. gulae and the subsequent activation of macrophages. Exposure of HEK cells transfected with Toll-like receptors (TLRs) or NOD-like receptors to P. gulae resulted in the ligation of TLR2, TLR4, and NOD2. The effects of this engagement of receptors were investigated by measuring the synthesis of nitric oxide (NO), CD86 expression, and inflammatory cytokine production by wild-type, TLR2-/-, and TLR4-/- macrophages. The addition of P. gulae to unprimed and gamma interferon (IFN-γ)-primed (M1 phenotype) macrophages significantly increased the surface expression of CD86, but only M1 macrophages produced nitric oxide. P. gulae-induced expression of CD86 on unprimed macrophages was dependent on both TLR2 and TLR4, but CD86 expression and NO production in M1 macrophages were only TLR2 dependent. P. gulae induced an increase in secretion of interleukin-1α (IL-1α), IL-1β, IL-6, IL-12p70, IL-13, tumor necrosis factor alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), monocyte chemoattractant protein 1 (MCP-1), and macrophage inflammatory protein 1α (MIP-1α) by M1 macrophages compared to that by unprimed controls. Among these cytokines, secretion of IL-6 and TNF-α by M1 macrophages was dependent on either TLR2 or TLR4. Our data indicate that TLR2 and TLR4 are important for P. gulae activation of unprimed macrophages and that activation and effector functions induced in M1 macrophages by P. gulae are mainly dependent on TLR2. In conclusion, P. gulae induces a strong TLR2-dependent inflammatory M1 macrophage response which may be important in establishing the chronic inflammation associated with periodontal disease in companion animals.
Collapse
|
110
|
Makarkov AI, Chierzi S, Pillet S, Murai KK, Landry N, Ward BJ. Plant-made virus-like particles bearing influenza hemagglutinin (HA) recapitulate early interactions of native influenza virions with human monocytes/macrophages. Vaccine 2017; 35:4629-4636. [DOI: 10.1016/j.vaccine.2017.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023]
|
111
|
The role of monocytes in models of infection by protozoan parasites. Mol Immunol 2017; 88:174-184. [PMID: 28704704 DOI: 10.1016/j.molimm.2017.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/29/2017] [Accepted: 06/04/2017] [Indexed: 02/07/2023]
Abstract
The confirmation of developmental differences between tissue macrophages and peripheral monocytes has changed our view of the functions and dynamics of these two important components of the innate immune system. It has been demonstrated conclusively that homeostasis of tissue resident macrophages is maintained by a low proliferative turn over. During an inflammatory response, bone marrow derived monocytes enter the tissue in large numbers and take part in the defense against the pathogens. After the destruction of invading pathogens, these cells disappear and tissue resident macrophages can be detected again. This new appreciation of the innate immune response has not only answered many outstanding questions regarding the role of the different myeloid cell types in inflammation, but also opened up new areas of research relating to the tissue- and pathogen-specific fate of the inflammatory macrophages or dendritic cells (DCs), and the transfer of this knowledge from mouse models to the human immune system. Nevertheless, there is still confusion in infection models, and especially in studies of human infections, as to what extent these recent observations and findings influence previous interpretations of data. This review will focus on insights from mouse models, summarize the literature on the ontogeny of macrophages and monocytes, explain the role of frequently used monocyte markers and effector molecules, and finally, discuss the role of inflammatory monocytes/macrophages/DCs in two experimental parasitic diseases.
Collapse
|
112
|
Mukherjee S, Subramaniam R, Chen H, Smith A, Keshava S, Shams H. Boosting efferocytosis in alveolar space using BCG vaccine to protect host against influenza pneumonia. PLoS One 2017; 12:e0180143. [PMID: 28686604 PMCID: PMC5501455 DOI: 10.1371/journal.pone.0180143] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/09/2017] [Indexed: 01/04/2023] Open
Abstract
Efferocytosis by alveolar phagocytes (APs) is pivotal in maintenance of lung homeostasis. Increased efferocytosis by APs results in protection against lethal acute lung injury due to pulmonary infections whereas defective efferocytosis by APs results in chronic lung inflammation. In this report, we show that pulmonary delivery of Bacillus Calmette-Guerin (BCG) significantly enhances efferocytosis by APs. Increased efferocytosis by APs maintains lung homeostasis and protects mice against lethal influenza pneumonia. Intranasally treated wild type C57Bl/6 (WT) mice with BCG showed significant increase in APs efferocytosis in vivo compared to their PBS-treated counterparts. All BCG-treated WT mice survived lethal influenza A virus (IAV) infection whereas all PBS-treated mice succumbed. BCG-induced resistance was abrogated by depleting AP prior to IAV infection. BCG treatment increased uptake, and digestion/removal of apoptotic cells by APs. BCG significantly increased the expression of TIM4 on APs and increased expression of Rab5 and Rab7. We demonstrated that increased efferocytosis by APs through pulmonary delivery of BCG initiated rapid clearance of apoptotic cells from the alveolar space, maintained lung homeostasis, reduced inflammation and protected host against lethal IAV pneumonia.
Collapse
Affiliation(s)
- Sanjay Mukherjee
- Center for Pulmonary and Infectious Diseases Control (CPIDC), University of Texas Health Science Center at Tyler, Tyler, TX, United States of America
| | - Renuka Subramaniam
- Center for Pulmonary and Infectious Diseases Control (CPIDC), University of Texas Health Science Center at Tyler, Tyler, TX, United States of America
| | - Han Chen
- Center for Pulmonary and Infectious Diseases Control (CPIDC), University of Texas Health Science Center at Tyler, Tyler, TX, United States of America
| | - Anthony Smith
- Center for Pulmonary and Infectious Diseases Control (CPIDC), University of Texas Health Science Center at Tyler, Tyler, TX, United States of America
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX, United States of America
| | - Homayoun Shams
- Center for Pulmonary and Infectious Diseases Control (CPIDC), University of Texas Health Science Center at Tyler, Tyler, TX, United States of America
- * E-mail:
| |
Collapse
|
113
|
Abstract
Monocytes and macrophages are professional phagocytes that occupy specific niches in every tissue of the body. Their survival, proliferation, and differentiation are controlled by signals from the macrophage colony-stimulating factor receptor (CSF-1R) and its two ligands, CSF-1 and interleukin-34. In this review, we address the developmental and transcriptional relationships between hematopoietic progenitor cells, blood monocytes, and tissue macrophages as well as the distinctions from dendritic cells. A huge repertoire of receptors allows monocytes, tissue-resident macrophages, or pathology-associated macrophages to adapt to specific microenvironments. These processes create a broad spectrum of macrophages with different functions and individual effector capacities. The production of large transcriptomic data sets in mouse, human, and other species provides new insights into the mechanisms that underlie macrophage functional plasticity.
Collapse
|
114
|
Abstract
In 1882, Elie Metchnikoff identified myeloid-like cells from starfish larvae responding to the invasion by a foreign body (rose thorn). This marked the origins for the study of innate immunity, and an appreciation that cellular immunity was well established even in these "primitive" organisms. This chapter focuses on these myeloid cells as well as the newest members of this family, the dendritic cells, and explores their evolutionary origins. Our goal is to provide evolutionary context for the development of the multilayered immune system of mammals, where myeloid cells now serve as central effectors of innate immunity and regulators of adaptive immunity. Overall, we find that core contributions of myeloid cells to the regulation of inflammation are based on mechanisms that have been honed over hundreds of millions of years of evolution. Using phagocytosis as a platform, we show how fairly simple beginnings have offered a robust foundation onto which additional control features have been integrated, resulting in central regulatory nodes that now manage multifactorial aspects of homeostasis and immunity.
Collapse
|
115
|
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that play a pivotal role in the pathogenesis of periodontitis. The use of animal models to study the role of DCs in periodontitis has been limited by lack of a method for sustained depletion of DCs. Hence, the objectives of this study were to validate the zDC-DTR knockin mouse model of conventional DCs (cDCs) depletion, as well as to investigate whether this depletion could be sustained long enough to induce alveolar bone loss in this model. zDC-DTR mice were treated with different dose regimens of diphtheria toxin (DT) to determine survival rate. A loading DT dose of 20ng/bw, followed and maintained with doses of 10ng/bm every 3days for up to 4weeks demonstrated 80% survival. Animals were weighed weekly and peripheral blood was obtained to confirm normal neutrophil counts. Five animals per group were euthanized at baseline, 24h, 1 and 4weeks. Bone marrow (BM), spleen (SP) and gingival tissue (GT) were harvested, and cells were isolated, separated and stained for Pre-DCs precursors (CD45R-MHCII+CD11c+Flt3+CD172a+) in BM, cDCs (CD11c+MHCII+CD209+) in spleen, and DCs in GT (CD45R+MHCII+CD11c+ DC-SIGN/CD209+). Pre-DCs in BM were significantly depleted at 24h and depletion maintained for up to 4weeks, as compared to blank (PBS) controls. Circulating cDCs in spleen demonstrated a non-significant trend to deplete in 1week with high variability among mice. GT also showed a similar non-significant trend to deplete in 24h. The zDC-DTR model seems to be viable for evaluating the role of DCs immune homeostasis disruption and alveolar bone loss pathogenesis in response to long-term oral infection.
Collapse
|
116
|
Cai Y, Shi Z, Bai Y. Review of Rosai-Dorfman Disease: New Insights into the Pathogenesis of This Rare Disorder. Acta Haematol 2017; 138:14-23. [PMID: 28614806 DOI: 10.1159/000475588] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/10/2017] [Indexed: 12/22/2022]
Abstract
Rosai-Dorfman disease (RDD) is a rare histiocytosis typically with bilateral painless cervical lymphadenopathy. Laboratory data are nonspecific, and the presence of emperipolesis in large foamy S-100+ CD1a- histiocytes is the prominent histologic feature. The pathogenesis of RDD still remains elusive. According to published studies, we propose that RDD cells might represent intermediate recruiting monocytes with differentiation blockade. Both disturbance of homoeostasis and inherent genomic alterations could contribute to initiation of the disorder through signal transduction. Several inflammatory molecules such as macrophage colony-stimulating factor, IL-1β, IL-6, and tumor necrosis factor-α also play a pivotal role in the development of this rare entity. Additional studies are needed to further elucidate the essence of the disease.
Collapse
Affiliation(s)
- Yanan Cai
- Department of Hematology/Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
| | | | | |
Collapse
|
117
|
Rahe MC, Murtaugh MP. Mechanisms of Adaptive Immunity to Porcine Reproductive and Respiratory Syndrome Virus. Viruses 2017; 9:v9060148. [PMID: 28608816 PMCID: PMC5490824 DOI: 10.3390/v9060148] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/25/2017] [Accepted: 06/07/2017] [Indexed: 02/07/2023] Open
Abstract
The adaptive immune response is necessary for the development of protective immunity against infectious diseases. Porcine reproductive and respiratory syndrome virus (PRRSV), a genetically heterogeneous and rapidly evolving RNA virus, is the most burdensome pathogen of swine health and wellbeing worldwide. Viral infection induces antigen-specific immunity that ultimately clears the infection. However, the resulting immune memory, induced by virulent or attenuated vaccine viruses, is inconsistently protective against diverse viral strains. The immunological mechanisms by which primary and memory protection are generated and used are not well understood. Here, we summarize current knowledge regarding cellular and humoral components of the adaptive immune response to PRRSV infection that mediate primary and memory immune protection against viruses.
Collapse
Affiliation(s)
- Michael C Rahe
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1971 Commonwealth Avenue, St. Paul, MN 55108, USA.
| | - Michael P Murtaugh
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 1971 Commonwealth Avenue, St. Paul, MN 55108, USA.
| |
Collapse
|
118
|
Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, Itzkovitz S, Colonna M, Schwartz M, Amit I. A Unique Microglia Type Associated with Restricting Development of Alzheimer's Disease. Cell 2017; 169:1276-1290.e17. [PMID: 28602351 DOI: 10.1016/j.cell.2017.05.018] [Citation(s) in RCA: 3287] [Impact Index Per Article: 410.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/10/2017] [Accepted: 05/11/2017] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) is a detrimental neurodegenerative disease with no effective treatments. Due to cellular heterogeneity, defining the roles of immune cell subsets in AD onset and progression has been challenging. Using transcriptional single-cell sorting, we comprehensively map all immune populations in wild-type and AD-transgenic (Tg-AD) mouse brains. We describe a novel microglia type associated with neurodegenerative diseases (DAM) and identify markers, spatial localization, and pathways associated with these cells. Immunohistochemical staining of mice and human brain slices shows DAM with intracellular/phagocytic Aβ particles. Single-cell analysis of DAM in Tg-AD and triggering receptor expressed on myeloid cells 2 (Trem2)-/- Tg-AD reveals that the DAM program is activated in a two-step process. Activation is initiated in a Trem2-independent manner that involves downregulation of microglia checkpoints, followed by activation of a Trem2-dependent program. This unique microglia-type has the potential to restrict neurodegeneration, which may have important implications for future treatment of AD and other neurodegenerative diseases. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Hadas Keren-Shaul
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Amit Spinrad
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Assaf Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), and University Medical Center, Cancer Genomics Netherlands, 3584 CG Utrecht, the Netherlands.
| | - Orit Matcovitch-Natan
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Raz Dvir-Szternfeld
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tyler K Ulland
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Kuti Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - David Lara-Astaiso
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Beata Toth
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
119
|
Splenic Ly6C hi monocytes contribute to adverse late post-ischemic left ventricular remodeling in heme oxygenase-1 deficient mice. Basic Res Cardiol 2017; 112:39. [PMID: 28534119 PMCID: PMC5440541 DOI: 10.1007/s00395-017-0629-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022]
Abstract
Heme oxygenase-1 (Hmox1) is a stress-inducible protein crucial in heme catabolism. The end products of its enzymatic activity possess anti-oxidative, anti-apoptotic and anti-inflammatory properties. Cardioprotective effects of Hmox1 were demonstrated in experimental models of myocardial infarction (MI). Nevertheless, its importance in timely resolution of post-ischemic inflammation remains incompletely understood. The aim of this study was to determine the role of Hmox1 in the monocyte/macrophage-mediated cardiac remodeling in a mouse model of MI. Hmox1 knockout (Hmox1-/-) and wild-type (WT, Hmox1+/+) mice were subjected to a permanent ligation of the left anterior descending coronary artery. Significantly lower incidence of left ventricle (LV) free wall rupture was noted between 3rd and 5th day after MI in Hmox1-/- mice resulting in their better overall survival. Then, starting from 7th until 21st day post-MI a more potent deterioration of LV function was observed in Hmox1-/- than in the surviving Hmox1+/+ mice. This was accompanied by higher numbers of Ly6Chi monocytes in peripheral blood, as well as higher expression of monocyte chemoattractant protein-1 and adhesion molecules in the hearts of MI-operated Hmox1-/- mice. Consequently, a greater post-MI monocyte-derived myocardial macrophage infiltration was noted in Hmox1-deficient individuals. Splenectomy decreased the numbers of circulating inflammatory Ly6Chi monocytes in blood, reduced the numbers of proinflammatory cardiac macrophages and significantly improved the post-MI LV function in Hmox1-/- mice. In conclusion, Hmox1 deficiency has divergent consequences in MI. On the one hand, it improves early post-MI survival by decreasing the occurrence of cardiac rupture. Afterwards, however, the hearts of Hmox1-deficient mice undergo adverse late LV remodeling due to overactive and prolonged post-ischemic inflammatory response. We identified spleen as an important source of these cardiovascular complications in Hmox1-/- mice.
Collapse
|
120
|
El Ghazal R, Yin X, Johns SC, Swanson L, Macal M, Ghosh P, Zuniga EI, Fuster MM. Glycan Sulfation Modulates Dendritic Cell Biology and Tumor Growth. Neoplasia 2017; 18:294-306. [PMID: 27237321 PMCID: PMC4887599 DOI: 10.1016/j.neo.2016.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/20/2022] Open
Abstract
In cancer, proteoglycans have been found to play roles in facilitating the actions of growth factors, and effecting matrix invasion and remodeling. However, little is known regarding the genetic and functional importance of glycan chains displayed by proteoglycans on dendritic cells (DCs) in cancer immunity. In lung carcinoma, among other solid tumors, tumor-associated DCs play largely subversive/suppressive roles, promoting tumor growth and progression. Herein, we show that targeting of DC glycan sulfation through mutation in the heparan sulfate biosynthetic enzyme N-deacetylase/N-sulfotransferase-1 (Ndst1) in mice increased DC maturation and inhibited trafficking of DCs to draining lymph nodes. Lymphatic-driven DC migration and chemokine (CCL21)-dependent activation of a major signaling pathway required for DC migration (as measured by phospho-Akt) were sensitive to Ndst1 mutation in DCs. Lewis lung carcinoma tumors in mice deficient in Ndst1 were reduced in size. Purified CD11c + cells from the tumors, which contain the tumor-infiltrating DC population, showed a similar phenotype in mutant cells. These features were replicated in mice deficient in syndecan-4, the major heparan sulfate proteoglycan expressed on the DC surface: Tumors were growth-impaired in syndecan-4–deficient mice and were characterized by increased infiltration by mature DCs. Tumors on the mutant background also showed greater infiltration by NK cells and NKT cells. These findings indicate the genetic importance of DC heparan sulfate proteoglycans in tumor growth and may guide therapeutic development of novel strategies to target syndecan-4 and heparan sulfate in cancer.
Collapse
Affiliation(s)
- Roland El Ghazal
- VA San Diego Healthcare System, Medical and Research Sections, La Jolla, CA 92161; Department of Medicine, Division of Pulmonary and Critical Care, University of California San Diego, La Jolla, CA 92037
| | - Xin Yin
- VA San Diego Healthcare System, Medical and Research Sections, La Jolla, CA 92161; Department of Medicine, Division of Pulmonary and Critical Care, University of California San Diego, La Jolla, CA 92037; Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Huaihai Institute of Technology, Lianyungang, China
| | - Scott C Johns
- VA San Diego Healthcare System, Medical and Research Sections, La Jolla, CA 92161; Department of Medicine, Division of Pulmonary and Critical Care, University of California San Diego, La Jolla, CA 92037
| | - Lee Swanson
- Division of Gastroenterology, University of California San Diego, La Jolla, CA 92093
| | - Monica Macal
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| | - Pradipta Ghosh
- Division of Gastroenterology, University of California San Diego, La Jolla, CA 92093
| | - Elina I Zuniga
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| | - Mark M Fuster
- VA San Diego Healthcare System, Medical and Research Sections, La Jolla, CA 92161; Department of Medicine, Division of Pulmonary and Critical Care, University of California San Diego, La Jolla, CA 92037; Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA 92093.
| |
Collapse
|
121
|
Abstract
Systemic administration of therapeutic agents has been the preferred approach to treat most pathological conditions, in particular for cancer therapy. This treatment modality is associated with side effects, off-target accumulation, toxicity, and rapid renal and hepatic clearance. Multiple efforts have focused on incorporating targeting moieties into systemic therapeutic vehicles to enhance retention and minimize clearance and side effects. However, only a small percentage of the nanoparticles administered systemically accumulate at the tumor site, leading to poor therapeutic efficacy. This has prompted researchers to call the status quo treatment regimen into question and to leverage new delivery materials and alternative administration routes to improve therapeutic outcomes. Recent approaches rely on the use of local delivery platforms that circumvent the hurdles of systemic delivery. Local administration allows delivery of higher "effective" doses while enhancing therapeutic molecules' stability, minimizing side effects, clearance, and accumulation in the liver and kidneys following systemic administration. Hydrogels have proven to be highly biocompatible materials that allow for versatile design to afford sensing and therapy at the same time. Hydrogels' chemical and physical versatility can be exploited to attain disease-triggered in situ assembly and hydrogel programmed degradation and consequent drug release, and hydrogels can also serve as a biocompatible depot for local delivery of stimuli-responsive therapeutic cargo. We will focus this Account on the hydrogel platform that we have developed in our lab, based on dendrimer amine and dextran aldehyde. This hydrogel is disease-responsive and capable of sensing the microenvironment and reacting in a graded manner to diverse pathologies to render different properties, including tissue adhesion, biocompatibility, hydrogel degradation, and embedded drug release profile. We also studied the degradation kinetics of our stimuli-responsive materials in vivo and analyzed the in vitro conditions under which in vitro-in vivo correlation is attained. Identifying key parameters in the in vivo microenvironment under healthy and disease conditions was key to attaining that correlation. The adhesive capacity of our dendrimer-dextran hydrogel makes it optimal for localized and sustained release of embedded drugs. We demonstrated that it affords the delivery of a range of therapeutics to combat cancer, including nucleic acids, small molecules, and antibody drugs. As a depot for local delivery, it allows a high dose of active biomolecules to be delivered directly at the tumor site. Immunotherapy, a recently blooming area in cancer therapy, may exploit stimuli-responsive hydrogels to impart systemic effects following localized therapy. Local delivery would enable release of the proper drug dose and improve drug bioavailability where needed at the same time creating memory and exerting the therapeutic effect systemically. This Account highlights our perspective on how local and systemic therapies provided by stimuli-responsive hydrogels should be used to impart more precise, long-lasting, and potent therapeutic outcomes.
Collapse
Affiliation(s)
- Nuria Oliva
- Department of Medicine,
Engineering in Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - João Conde
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- School of
Engineering and Materials Science, Queen Mary University of London, London E1 4NS, U.K
| | - Kui Wang
- Department of Medicine,
Engineering in Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Natalie Artzi
- Department of Medicine,
Engineering in Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02139, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
122
|
Marinkovic E, Djokic R, Lukic I, Filipovic A, Inic-Kanada A, Kosanovic D, Gavrovic-Jankulovic M, Stojanovic M. Modulation of functional characteristics of resident and thioglycollate-elicited peritoneal murine macrophages by a recombinant banana lectin. PLoS One 2017; 12:e0172469. [PMID: 28235050 PMCID: PMC5325268 DOI: 10.1371/journal.pone.0172469] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/06/2017] [Indexed: 01/25/2023] Open
Abstract
We demonstrated that a recombinant banana lectin (rBanLec), which structural characteristics and physiological impacts highly resemble those reported for its natural counterparts, binds murine peritoneal macrophages and specifically modulates their functional characteristics. By using rBanLec in concentrations ranging from 1 μg to 10 μg to stimulate resident (RMs) and thioglycollate-elicited (TGMs) peritoneal macrophages from BALB/c and C57BL/6 mice, we have shown that effects of rBanLec stimulation depend on its concentration but also on the functional status of macrophages and their genetic background. rBanLec, in a positive dose-dependent manner, promotes the proliferation of TGMs from both BALB/c and C57BL/6 mice, while its mitogenic influence on RMs is significantly lower (BALB/c mice) or not detectable (C57BL/6 mice). In all peritoneal macrophages, irrespective of their type and genetic background, rBanLec, in a positive dose dependent manner, enhances the secretion of IL-10. rBanLec stimulation of RMs from both BALB/c and C57BL/6 resulted in a positive dose-dependent promotion of proinflammatory phenotype (enhancement of NO production and IL-12 and TNFα secretion, reduction of arginase activity). Positive dose-dependent skewing toward proinflammatory phenotype was also observed in TGMs from C57BL/6 mice. However, the enhancement of rBanLec stimulation promotes skewing of TGMs from BALB/c mice towards anti-inflammatory profile (reduction of NO production and IL-12 secretion, enhancement of arginase activity and TGFβ and IL-4 secretion). Moreover, we established that rBanLec binds oligosaccharide structures of TLR2 and CD14 and that blocking of signaling via these receptors significantly impairs the production of TNFα and NO in BALB/c macrophages. Since the outcome of rBanLec stimulation depends on rBanLec concentration as well as on the functional characteristics of its target cells and their genetic background, further studies are needed to investigate its effects under physiological and specific pathological conditions.
Collapse
Affiliation(s)
- Emilija Marinkovic
- Department of Research and Development; Institute of Virology, Vaccines and Sera–TORLAK; Belgrade, Serbia
| | - Radmila Djokic
- Department of Research and Development; Institute of Virology, Vaccines and Sera–TORLAK; Belgrade, Serbia
| | - Ivana Lukic
- Department of Research and Development; Institute of Virology, Vaccines and Sera–TORLAK; Belgrade, Serbia
| | - Ana Filipovic
- Department of Research and Development; Institute of Virology, Vaccines and Sera–TORLAK; Belgrade, Serbia
| | - Aleksandra Inic-Kanada
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centres of Expertise; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna, Austria
| | - Dejana Kosanovic
- Department of Research and Development; Institute of Virology, Vaccines and Sera–TORLAK; Belgrade, Serbia
| | | | - Marijana Stojanovic
- Department of Research and Development; Institute of Virology, Vaccines and Sera–TORLAK; Belgrade, Serbia
| |
Collapse
|
123
|
Rheb1 deletion in myeloid cells aggravates OVA-induced allergic inflammation in mice. Sci Rep 2017; 7:42655. [PMID: 28225024 PMCID: PMC5320517 DOI: 10.1038/srep42655] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 01/12/2017] [Indexed: 12/28/2022] Open
Abstract
The small GTPase ras homolog enriched in brain (Rheb) is a downstream target of tuberous sclerosis complex 1/2 (TSC1/2) and an upstream activator of the mechanistic target of rapamycin complex 1 (mTORC1), the emerging essential modulator of M1/M2 balance in macrophages. However, the role and regulatory mechanisms of Rheb in macrophage polarization and allergic asthma are not known. In the present study, we utilized a mouse model with myeloid cell-specific deletion of the Rheb1 gene and an ovalbumin (OVA)-induced allergic asthma model to investigate the role of Rheb1 in allergic asthma and macrophage polarization. Increased activity of Rheb1 and mTORC1 was observed in myeloid cells of C57BL/6 mice with OVA-induced asthma. In an OVA-induced asthma model, Rheb1-KO mice demonstrated a more serious inflammatory response, more mucus production, enhanced airway hyper-responsiveness, and greater eosinophil numbers in bronchoalveolar lavage fluid (BALF). They also showed increased numbers of bone marrow macrophages and BALF myeloid cells, elevated M2 polarization and reduced M1 polarization of macrophages. Thus, we have established that Rheb1 is critical for the polarization of macrophages and inhibition of allergic asthma. Deletion of Rheb1 enhances M2 polarization but decreases M1 polarization in alveolar macrophages, leading to the aggravation of OVA-induced allergic asthma.
Collapse
|
124
|
van der Spek AH, Fliers E, Boelen A. Thyroid hormone metabolism in innate immune cells. J Endocrinol 2017; 232:R67-R81. [PMID: 27852725 DOI: 10.1530/joe-16-0462] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/13/2016] [Indexed: 12/23/2022]
Abstract
Thyroid hormone (TH) metabolism and thyroid status have been linked to various aspects of the immune response. There is extensive literature available on the effects of thyroid hormone on innate immune cells. However, only recently have authors begun to study the mechanisms behind these effects and the role of intracellular TH metabolism in innate immune cell function during inflammation. This review provides an overview of the molecular machinery of intracellular TH metabolism present in neutrophils, macrophages and dendritic cells and the role and effects of intracellular TH metabolism in these cells. Circulating TH levels have a profound effect on neutrophil, macrophage and dendritic cell function. In general, increased TH levels result in an amplification of the pro-inflammatory response of these cells. The mechanisms behind these effects include both genomic and non-genomic effects of TH. Besides a pro-inflammatory effect induced by extracellular TH, the cellular response to pro-inflammatory stimuli appears to be dependent on functional intracellular TH metabolism. This is illustrated by the fact that the deiodinase enzymes and in some cell types also thyroid hormone receptors appear to be crucial for adequate innate immune cell function. This overview of the literature suggests that TH metabolism plays an important role in the host defence against infection through the modulation of innate immune cell function.
Collapse
Affiliation(s)
- Anne H van der Spek
- Department of Endocrinology and MetabolismAcademic Medical Center, Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and MetabolismAcademic Medical Center, Amsterdam, The Netherlands
| | - Anita Boelen
- Department of Endocrinology and MetabolismAcademic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
125
|
George JF, Lever JM, Agarwal A. Mononuclear phagocyte subpopulations in the mouse kidney. Am J Physiol Renal Physiol 2017; 312:F640-F646. [PMID: 28100500 DOI: 10.1152/ajprenal.00369.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 12/13/2022] Open
Abstract
Mononuclear phagocytes are the most common cells in the kidney associated with immunity and inflammation. Although the presence of these cells in the kidney has been known for decades, the study of mononuclear phagocytes in the context of kidney function and dysfunction is still at an early stage. The purpose of this review is to summarize the present knowledge regarding classification of these cells in the mouse kidney and to identify relevant questions that would further advance the field and potentially lead to new opportunities for treatment of acute kidney injury and other kidney diseases.
Collapse
Affiliation(s)
- James F George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Nephrology Research and Training Center; University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Jeremie M Lever
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Nephrology Research and Training Center; University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; .,Department of Nephrology Research and Training Center; University of Alabama at Birmingham, Birmingham, Alabama; and.,Department of Veterans Affairs, Birmingham, Alabama
| |
Collapse
|
126
|
Gao H, Danzi MC, Choi CS, Taherian M, Dalby-Hansen C, Ellman DG, Madsen PM, Bixby JL, Lemmon VP, Lambertsen KL, Brambilla R. Opposing Functions of Microglial and Macrophagic TNFR2 in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. Cell Rep 2017; 18:198-212. [PMID: 28052249 PMCID: PMC5218601 DOI: 10.1016/j.celrep.2016.11.083] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/30/2016] [Accepted: 11/30/2016] [Indexed: 12/15/2022] Open
Abstract
In multiple sclerosis (MS), soluble tumor necrosis factor (TNF) is detrimental via activation of TNF receptor 1 (TNFR1), whereas transmembrane TNF is beneficial primarily by activating TNF receptor 2 (TNFR2). Here, we investigate the role of TNFR2 in microglia and monocytes/macrophages in experimental autoimmune encephalomyelitis (EAE), a model of MS, by cell-specific gene targeting. We show that TNFR2 ablation in microglia leads to early onset of EAE with increased leukocyte infiltration, T cell activation, and demyelination in the central nervous system (CNS). Conversely, TNFR2 ablation in monocytes/macrophages results in EAE suppression with impaired peripheral T cell activation and reduced CNS T cell infiltration and demyelination. Our work uncovers a dichotomy of function for TNFR2 in myeloid cells, with microglial TNFR2 providing protective signals to contain disease and monocyte/macrophagic TNFR2 driving immune activation and EAE initiation. This must be taken into account when targeting TNFR2 for therapeutic purposes in neuroinflammatory diseases.
Collapse
MESH Headings
- Animals
- CX3C Chemokine Receptor 1/metabolism
- Cell Proliferation
- Chronic Disease
- Demyelinating Diseases/genetics
- Demyelinating Diseases/metabolism
- Demyelinating Diseases/pathology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Deletion
- Gene Expression Regulation
- Homeostasis/genetics
- Inflammation/pathology
- Macrophages/metabolism
- Mice, Inbred C57BL
- Microglia/metabolism
- Myelin Sheath/metabolism
- Neuroprotection
- Phenotype
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Sequence Analysis, RNA
- Spinal Cord/pathology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- Transcriptome/genetics
Collapse
Affiliation(s)
- Han Gao
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matt C Danzi
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Mehran Taherian
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Camilla Dalby-Hansen
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C 5000, Denmark
| | - Ditte G Ellman
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C 5000, Denmark
| | - Pernille M Madsen
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C 5000, Denmark
| | - John L Bixby
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Cellular and Molecular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vance P Lemmon
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kate L Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C 5000, Denmark; Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C 5000, Denmark; Department of Neurology, Odense University Hospital, Odense C 5000, Denmark
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
127
|
Application of Immunohistochemistry in Toxicologic Pathology of the Hematolymphoid System. IMMUNOPATHOLOGY IN TOXICOLOGY AND DRUG DEVELOPMENT 2017. [DOI: 10.1007/978-3-319-47377-2_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
128
|
Siewe N, Yakubu AA, Satoskar AR, Friedman A. Granuloma formation in leishmaniasis: A mathematical model. J Theor Biol 2017; 412:48-60. [DOI: 10.1016/j.jtbi.2016.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 10/03/2016] [Accepted: 10/14/2016] [Indexed: 12/26/2022]
|
129
|
Italiani P, Boraschi D. Development and Functional Differentiation of Tissue-Resident Versus Monocyte-Derived Macrophages in Inflammatory Reactions. Results Probl Cell Differ 2017; 62:23-43. [PMID: 28455704 DOI: 10.1007/978-3-319-54090-0_2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Mononuclear phagocytes are key cells in tissue integrity and defense. Tissue-resident macrophages are abundantly present in all tissues of the body and have a complex role in ensuring tissue functions and homeostatic balance. Circulating blood monocytes can enter tissue both in steady-state conditions, for helping in replenishing the tissue-resident macrophage pool and, in particular, for acting as potent effector cells during inflammatory events such as infections, traumas, and diseases. The heterogeneity of monocytes and macrophages depends on their ontogeny, their tissue location, and their functional programming, with both monocytes and macrophages able to exert distinct or similar functions depending on the tissue-specific and stimulus-specific microenvironment. In this short review, we will review the current hypotheses on tissue-resident macrophage ontogeny and functions, as compared to blood-derived monocytes, with a particular focus on inflammatory conditions.
Collapse
Affiliation(s)
- Paola Italiani
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Diana Boraschi
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| |
Collapse
|
130
|
Abstract
Macrophages are found in all tissues and regulate tissue morphogenesis during development through trophic and scavenger functions. The colony stimulating factor-1 (CSF-1) receptor (CSF-1R) is the major regulator of tissue macrophage development and maintenance. In combination with receptor activator of nuclear factor κB (RANK), the CSF-1R also regulates the differentiation of the bone-resorbing osteoclast and controls bone remodeling during embryonic and early postnatal development. CSF-1R-regulated macrophages play trophic and remodeling roles in development. Outside the mononuclear phagocytic system, the CSF-1R directly regulates neuronal survival and differentiation, the development of intestinal Paneth cells and of preimplantation embryos, as well as trophoblast innate immune function. Consistent with the pleiotropic roles of the receptor during development, CSF-1R deficiency in most mouse strains causes embryonic or perinatal death and the surviving mice exhibit multiple developmental and functional deficits. The CSF-1R is activated by two dimeric glycoprotein ligands, CSF-1, and interleukin-34 (IL-34). Homozygous Csf1-null mutations phenocopy most of the deficits of Csf1r-null mice. In contrast, Il34-null mice have no gross phenotype, except for decreased numbers of Langerhans cells and microglia, indicating that CSF-1 plays the major developmental role. Homozygous inactivating mutations of the Csf1r or its ligands have not been reported in man. However, heterozygous inactivating mutations in the Csf1r lead to a dominantly inherited adult-onset progressive dementia, highlighting the importance of CSF-1R signaling in the brain.
Collapse
Affiliation(s)
- Violeta Chitu
- Albert Einstein College of Medicine, Bronx, NY, United States
| | | |
Collapse
|
131
|
Chen GY, Li Z, Duarte JN, Esteban A, Cheloha RW, Theile CS, Fink GR, Ploegh HL. Rapid capture and labeling of cells on single domain antibodies-functionalized flow cell. Biosens Bioelectron 2016; 89:789-794. [PMID: 27816596 DOI: 10.1016/j.bios.2016.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/01/2016] [Accepted: 10/05/2016] [Indexed: 01/13/2023]
Abstract
Current techniques to characterize leukocyte subgroups in blood require long sample preparation times and sizable sample volumes. A simplified method for leukocyte characterization using smaller blood volumes would thus be useful in diagnostic settings. Here we describe a flow system comprised of two functionalized graphene oxide (GO) surfaces that allow the capture of distinct leukocyte populations from small volumes blood using camelid single-domain antibodyfragments (VHHs) as capture agents. We used site-specifically labeled leukocytes to detect and identify cells exposed to fungal challenge. Combining the chemical and optical properties of GO with the versatility of the VHH scaffold in the context of a flow system provides a quick and efficient method for the capture and characterization of functional leukocytes.
Collapse
Affiliation(s)
- Guan-Yu Chen
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
| | - Zeyang Li
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Joao N Duarte
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Ross W Cheloha
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Gerald R Fink
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
132
|
Klein B, Haggeney T, Fietz D, Indumathy S, Loveland KL, Hedger M, Kliesch S, Weidner W, Bergmann M, Schuppe HC. Specific immune cell and cytokine characteristics of human testicular germ cell neoplasia. Hum Reprod 2016; 31:2192-202. [PMID: 27609978 DOI: 10.1093/humrep/dew211] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/22/2016] [Indexed: 12/16/2022] Open
Abstract
STUDY QUESTION Which immune cells and cytokine profiles are characteristic for testicular germ cell neoplasia and what consequences does this have for the understanding of the related testicular immunopathology? SUMMARY ANSWER The unique immune environment of testicular germ cell neoplasia comprises B cells and dendritic cells as well as high transcript levels of IL-6 and other B cell supporting or T helper cell type 1 (Th1)-driven cytokines and thus differs profoundly from normal testis or inflammatory lesions associated with hypospermatogenesis. WHAT IS KNOWN ALREADY T cells are known to be the major component of inflammatory infiltrates associated with either hypospermatogenesis or testicular cancer. It has previously been reported that B cells are only involved within infiltrates of seminoma samples, but this has not been investigated further. STUDY DESIGN, SIZE, DURATION Immunohistochemical characterisation (IHC) of infiltrating immune cells and RT-qPCR-based analysis of corresponding cytokine microenvironments was performed on different testicular pathologies. Testicular biopsies, obtained from men undergoing andrological work-up of infertility or taken during surgery for testicular cancer, were used in this study. Samples were grouped as follows: (i) normal spermatogenesis (n = 18), (ii) hypospermatogenesis associated with lymphocytic infiltrates (n = 10), (iii) samples showing neoplasia [germ cell neoplasia in situ (GCNIS, n = 26) and seminoma, n = 18]. PARTICIPANTS/MATERIALS, SETTING, METHODS IHC was performed using antibodies against T cells (CD3+), B cells (CD20cy+), dendritic cells (CD11c+), macrophages (CD68+) and mast cells (mast cell tryptase+). Degree and compartmental localisation of immune cells throughout all groups analysed was evaluated semi-quantitatively. RT-qPCR on RNA extracted from cryo-preserved tissue samples was performed to analyse mRNA cytokine expression, specifically levels of IL-1β, IL-6, IL-17a, tumour necrosis factor (TNF)-α (pro-inflammatory), IL-10, transforming growth factor (TGF)-β1 (anti-inflammatory), IL-2, IL-12a, IL-12b, interferon (IFN)-γ (Th1-driven), IL-4, IL-5, IL-13, IL-23a (Th2-driven), CXCL-13, CXCL-10 and CCL-5 (chemokines). MAIN RESULTS AND THE ROLE OF CHANCE This is the first study showing a direct linkage between the distribution pattern of immune cells in hypospermatogenesis versus testicular cancer and analysis of a wide range of 17 related cyto- and chemokines. A fundamental difference between testicular inflammation patterns associated with different testicular inflammatory conditions either containing or lacking neoplastic cells was demonstrated. In hypospermatogenesis, T cells were detected, whereas B cells and dendritic cells were almost absent. Within GCNIS and seminoma, in addition to T cells, high numbers of dendritic cells and B cells were found, the latter additionally organised in cell clusters, whereas mast cells were absent. Transcripts encoding pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α), anti-inflammatory cytokines (TGF-β1), Th1-driven cytokines (IL-2 and IFN-γ) as well as chemokines (CXCL-13, CXCL-10 and CCL-5) were all significantly increased in testicular germ cell neoplasia (P ≤ 0.01), suggesting the presence of a pro-tumorigenic environment. In contrast, Th2-related cytokines (IL-5, IL-13 and IL-23a) characterised the environment within samples showing normal spermatogenesis as well as hypospermatogenesis. One of the most important outcomes is the pivotal role of IL-6 in testicular cancer that opens potential novel diagnostic and/or immune-therapeutic perspective for testis cancer. LIMITATIONS, REASONS FOR CAUTION Testicular tissue composed of immune cells as well as other somatic cells and germ cells does not allow identification of specific cytokine sources or single cell types, being responsible for establishing the overall cytokine environment. In this study, laser-assisted microdissection did not reach the required efficiency for RT-qPCR analyses. Therefore, in vitro models would be suggested for addressing the above-mentioned issue. Conclusions about cytokine levels in testes with GCNIS are based on a small number of samples. WIDER IMPLICATIONS OF THE FINDINGS The unique B cell presence and the significantly increased expression level of IL-6 in testicular germ cell neoplasia (P < 0.001) strengthen its special role in this disease. In line with current knowledge on other types of cancer, these results underline the relevance of further investigating the potential of IL-6 as early biomarker and target for therapeutic intervention in testicular germ cell neoplasia. STUDY FUNDING/COMPETING INTERESTS This study (and B.K. in person) was supported by the Deutsche Forschungsgemeinschaft (DFG) as part of the International Research Training Group between Justus Liebig University of Giessen and Monash University, Melbourne (GRK 1871/1) on 'Molecular pathogenesis on male reproductive disorders'. T.H., H.-C.S. and M.B. were supported by the LOEWE focus group 'MIBIE' (male infertility during infection & inflammation)-an excellence initiative of the German state government of Hessen. From the Australian side, K.L. was supported by NHMRC grants (Fellowship, ID1079646 and Project, ID1081987); K.L., S.I. and M.H. received scholarship (S.I.) and research funding (K.L., M.H.) from Monash University. The project also drew support from the Victorian Government's Operational Infrastructure Support Program. The authors have no competing interests to declare.
Collapse
Affiliation(s)
- Britta Klein
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Frankfurter Strasse 98, 35392 Giessen, Germany Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Thomas Haggeney
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Frankfurter Strasse 98, 35392 Giessen, Germany
| | - Daniela Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Frankfurter Strasse 98, 35392 Giessen, Germany
| | - Sivanjah Indumathy
- Hudson Institute of Medical Research, Wright Street, Clayton, VIC 3168, Australia
| | - Kate L Loveland
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia Hudson Institute of Medical Research, Wright Street, Clayton, VIC 3168, Australia School of Clinical Sciences, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Mark Hedger
- Hudson Institute of Medical Research, Wright Street, Clayton, VIC 3168, Australia
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University of Muenster, Domagkstrasse 11, 48129 Muenster, Germany
| | - Wolfgang Weidner
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Rudolf-Buchheim Str. 7, 35392 Giessen, Germany
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Frankfurter Strasse 98, 35392 Giessen, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Rudolf-Buchheim Str. 7, 35392 Giessen, Germany
| |
Collapse
|
133
|
Rotavirus Recombinant VP6 Nanotubes Act as an Immunomodulator and Delivery Vehicle for Norovirus Virus-Like Particles. J Immunol Res 2016; 2016:9171632. [PMID: 27689099 PMCID: PMC5027051 DOI: 10.1155/2016/9171632] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 07/05/2016] [Accepted: 07/19/2016] [Indexed: 01/26/2023] Open
Abstract
We have recently shown that tubular form of rotavirus (RV) recombinant VP6 protein has an in vivo adjuvant effect on the immunogenicity of norovirus (NoV) virus-like particle (VLP) vaccine candidate. In here, we investigated in vitro effect of VP6 on antigen presenting cell (APC) activation and maturation and whether VP6 facilitates NoV VLP uptake by these APCs. Mouse macrophage cell line RAW 264.7 and dendritic cell line JAWSII were used as model APCs. Internalization of VP6, cell surface expression of CD40, CD80, CD86, and major histocompatibility class II molecules, and cytokine and chemokine production were analyzed. VP6 nanotubes were efficiently internalized by APCs. VP6 upregulated the expression of cell surface activation and maturation molecules and induced secretion of several proinflammatory cytokines and chemokines. The mechanism of VP6 action was shown to be partially dependent on lipid raft-mediated endocytic pathway as shown by methyl-β-cyclodextrin inhibition on tumor necrosis factor α secretion. These findings add to the understanding of mechanism by which VP6 exerts its immunostimulatory and immunomodulatory actions and further support its use as a part of nonlive RV-NoV combination vaccine.
Collapse
|
134
|
Papadopoulos G, Shaik-Dasthagirisaheb YB, Huang N, Viglianti GA, Henderson AJ, Kantarci A, Gibson FC. Immunologic environment influences macrophage response to Porphyromonas gingivalis. Mol Oral Microbiol 2016; 32:250-261. [PMID: 27346827 DOI: 10.1111/omi.12168] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2016] [Indexed: 02/03/2023]
Abstract
Macrophages adapt both phenotypically and functionally to the cytokine balance in host tissue microenvironments. Recent studies established that macrophages contribute an important yet poorly understood role in the development of infection-elicited oral bone loss. We hypothesized that macrophage adaptation to inflammatory signals encountered before pathogen interaction would significantly influence the subsequent immune response of these cells to the keystone oral pathobiont Porphyromonas gingivalis. Employing classically activated (M1) and alternatively activated (M2) murine bone-marrow-derived macrophage (BMDMø), we observed that immunologic activation of macrophages before P. gingivalis challenge dictated phenotype-specific changes in the expression of inflammation-associated molecules important to sensing and tuning host response to bacterial infection including Toll-like receptors 2 and 4, CD14, CD18 and CD11b (together comprising CR3), major histocompatibility complex class II, CD80, and CD86. M2 cells responded to P. gingivalis with higher expression of tumor necrosis factor-α, interleukin-6, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α, regulated on activation normal T cell expressed and secreted, and KC than M1 cells. M1 BMDMø expressed higher levels of interleukin-10 to P. gingivalis than M2 BMDMø. Functionally, we observed that M2 BMDMø bound P. gingivalis more robustly than M1 BMDMø. These data describe an important contribution of macrophage skewing in the subsequent development of the cellular immune response to P. gingivalis.
Collapse
Affiliation(s)
- G Papadopoulos
- Section of infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Y B Shaik-Dasthagirisaheb
- Section of infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - N Huang
- Section of infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - G A Viglianti
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - A J Henderson
- Section of infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - A Kantarci
- Department of Applied Oral Sciences, Forsyth Institute, Cambridge, MA, USA
| | - F C Gibson
- Section of infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
135
|
Broadley S, Plaumann A, Coletti R, Lehmann C, Wanisch A, Seidlmeier A, Esser K, Luo S, Rämer P, Massberg S, Busch D, van Lookeren Campagne M, Verschoor A. Dual-Track Clearance of Circulating Bacteria Balances Rapid Restoration of Blood Sterility with Induction of Adaptive Immunity. Cell Host Microbe 2016; 20:36-48. [DOI: 10.1016/j.chom.2016.05.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/15/2016] [Accepted: 05/26/2016] [Indexed: 12/25/2022]
|
136
|
Unanue ER, Ferris ST, Carrero JA. The role of islet antigen presenting cells and the presentation of insulin in the initiation of autoimmune diabetes in the NOD mouse. Immunol Rev 2016; 272:183-201. [PMID: 27319351 PMCID: PMC4938008 DOI: 10.1111/imr.12430] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We have been examining antigen presentation and the antigen presenting cells (APCs) in the islets of Langerhans of the non-obese diabetic (NOD) mouse. The purpose is to identify the earliest events that initiate autoimmunity in this confined tissue. Islets normally have a population of macrophages that is distinct from those that inhabit the exocrine pancreas. Also found in NOD islets is a minor population of dendritic cells (DCs) that bear the CD103 integrin. We find close interactions between beta cells and the two APCs that result in the initiation of the autoimmunity. Even under non-inflammatory conditions, beta cells transfer insulin-containing vesicles to the APCs of the islet. This reaction requires live cells and intimate contact. The autoimmune process starts in islets with the entrance of CD4(+) T cells and an increase in the CD103(+) DCs. Mice deficient in the Batf3 transcription factor never develop diabetes due to the absence of the CD103/CD8α lineage of DCs. We hypothesize that the 12-20 peptide of the beta chain of insulin is responsible for activation of the initial CD4(+) T-cell response during diabetogenesis.
Collapse
Affiliation(s)
- Emil R. Unanue
- Department of Pathology and Immunology, Division of Immunobiology, 660 South Euclid Avenue, Campus Box 8118, Washington University School of Medicine, St. Louis, Missouri USA 63110
| | - Stephen T. Ferris
- Department of Pathology and Immunology, Division of Immunobiology, 660 South Euclid Avenue, Campus Box 8118, Washington University School of Medicine, St. Louis, Missouri USA 63110
| | - Javier A. Carrero
- Department of Pathology and Immunology, Division of Immunobiology, 660 South Euclid Avenue, Campus Box 8118, Washington University School of Medicine, St. Louis, Missouri USA 63110
| |
Collapse
|
137
|
Katsura Y, Harada N, Harada S, Ishimori A, Makino F, Ito J, Kamachi F, Okumura K, Akiba H, Atsuta R, Takahashi K. Characteristics of alveolar macrophages from murine models of OVA-induced allergic airway inflammation and LPS-induced acute airway inflammation. Exp Lung Res 2016; 41:370-82. [PMID: 26151756 DOI: 10.3109/01902148.2015.1044137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Macrophages include the classically activated pro-inflammatory M1 macrophages (M1s) and alternatively activated anti-inflammatory M2 macrophages (M2s). The M1s are activated by both interferon-γ and Toll-like receptor ligands, including lipopolysaccharide (LPS), and have potent pro-inflammatory activity. In contrast, Th2 cytokines activate the M2s, which are involved in the immune response to parasites, promotion of tissue remodeling, and immune regulatory functions. Although alveolar macrophages (AMs) play an essential role in the pulmonary immune system, little is known about their phenotypes. METHODS Quantitative reverse transcription polymerase chain reaction and flow cytometry were used to define the characteristics of alveolar macrophages derived from untreated naïve mice and from murine models of both ovalbumin (OVA)-induced allergic airway inflammation and LPS-induced acute airway inflammation. AMs were co-cultured with CD4(+) T cells and were pulsed with tritiated thymidine to assess proliferative responses. RESULTS We characterized in detail murine AMs and found that these cells were not completely consistent with the current M1 versus M2-polarization model. OVA-induced allergic and LPS-induced acute airway inflammation promoted the polarization of AMs towards the current M2-skewed and M1-skewed phenotypes, respectively. Moreover, our data also show that CD11c(+) CD11b(+) AMs from the LPS-treated mice play a regulatory role in antigen-specific T-cell proliferation in vitro. CONCLUSIONS These characteristics of AMs depend on the incoming pathogens they encounter and on the phase of inflammation and do not correspond to the current M1 versus M2-polarization model. These findings may facilitate an understanding of their contributions to the pulmonary immune system in airway inflammation.
Collapse
Affiliation(s)
- Yoko Katsura
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan.,b 2 Research Institute for Diseases of Old Ages , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan.,c 3 Department of Immunology , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Norihiro Harada
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan.,b 2 Research Institute for Diseases of Old Ages , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan.,c 3 Department of Immunology , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Sonoko Harada
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan.,c 3 Department of Immunology , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Ayako Ishimori
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Fumihiko Makino
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Jun Ito
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Fumitaka Kamachi
- c 3 Department of Immunology , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Ko Okumura
- d 4 Atopy (Allergy) Research Center , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Hisaya Akiba
- c 3 Department of Immunology , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Ryo Atsuta
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| | - Kazuhisa Takahashi
- a 1 Department of Respiratory Medicine , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan.,b 2 Research Institute for Diseases of Old Ages , Juntendo University Faculty of Medicine and Graduate School of Medicine , Tokyo, Japan
| |
Collapse
|
138
|
Dando SJ, Naranjo Golborne C, Chinnery HR, Ruitenberg MJ, McMenamin PG. A case of mistaken identity: CD11c-eYFP(+) cells in the normal mouse brain parenchyma and neural retina display the phenotype of microglia, not dendritic cells. Glia 2016; 64:1331-49. [PMID: 27189804 DOI: 10.1002/glia.23005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Under steady-state conditions the central nervous system (CNS) is traditionally thought to be devoid of antigen presenting cells; however, putative dendritic cells (DCs) expressing enhanced yellow fluorescent protein (eYFP) are present in the retina and brain parenchyma of CD11c-eYFP mice. We previously showed that these mice carry the Crb1(rd8) mutation, which causes retinal dystrophic lesions; therefore we hypothesized that the presence of CD11c-eYFP(+) cells within the CNS may be due to pathology associated with the Crb1(rd8) mutation. We generated CD11c-eYFP Crb1(wt/wt) mice and compared the distribution and immunophenotype of CD11c-eYFP(+) cells in CD11c-eYFP mice with and without the Crb1(rd8) mutation. The number and distribution of CD11c-eYFP(+) cells in the CNS was similar between CD11c-eYFP Crb1(wt/wt) and CD11c-eYFP Crb1(rd8/rd8) mice. CD11c-eYFP(+) cells were distributed throughout the inner retina, and clustered in brain regions that receive input from the external environment or lack a blood-brain barrier. CD11c-eYFP(+) cells within the retina and cerebral cortex of CD11c-eYFP Crb1(wt/wt) mice expressed CD11b, F4/80, CD115 and Iba-1, but not DC or antigen presentation markers, whereas CD11c-eYFP(+) cells within the choroid plexus and pia mater expressed CD11c, I-A/I-E, CD80, CD86, CD103, DEC205, CD8α and CD135. The immunophenotype of CD11c-eYFP(+) cells and microglia within the CNS was similar between CD11c-eYFP Crb1(wt/wt) and CD11c-eYFP Crb1(rd8/rd8) mice; however, CD11c and I-A/I-E expression was significantly increased in CD11c-eYFP Crb1(rd8/rd8) mice. This study demonstrates that the overwhelming majority of CNS CD11c-eYFP(+) cells do not display the phenotype of DCs or their precursors and are most likely a subpopulation of microglia. GLIA 2016. GLIA 2016;64:1331-1349.
Collapse
Affiliation(s)
- Samantha J Dando
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Cecilia Naranjo Golborne
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, the University of Queensland, Brisbane, Queensland, Australia.,Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Paul G McMenamin
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
139
|
Zhang Y, Li JQ, Jiang ZZ, Li L, Wu Y, Zheng L. CD169 identifies an anti-tumour macrophage subpopulation in human hepatocellular carcinoma. J Pathol 2016; 239:231-41. [DOI: 10.1002/path.4720] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/21/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yi Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center, Sun Yat-sen University; Guangzhou PR China
| | - Jin-Qing Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center, Sun Yat-sen University; Guangzhou PR China
| | - Ze-Zhou Jiang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
| | - Lian Li
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
| | - Yan Wu
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
| | - Limin Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center, Sun Yat-sen University; Guangzhou PR China
| |
Collapse
|
140
|
Siewe N, Yakubu AA, Satoskar AR, Friedman A. Immune response to infection by Leishmania: A mathematical model. Math Biosci 2016; 276:28-43. [PMID: 26987853 DOI: 10.1016/j.mbs.2016.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 10/22/2022]
Abstract
Leishmaniasis is a disease caused by the Leishmania parasites. The injection of the parasites into the host occurs when a sand fly, which is the vector, bites the skin of the host. The parasites, which are obligate, take advantage of the immune system response and invade both the classically activated macrophages (M1) and the alternatively activated macrophages (M2). In this paper, we develop a mathematical model to explain the evolution of the disease. Simulations of the model show that, M2 macrophages steadily increase and M1 macrophages steadily decrease, while M1+M2 reach a steady state which is approximately the same as at healthy state of the host. Furthermore, the ratio of Leishmania parasites to macrophages depends homogeneously on their ratio at the time of the initial infection, in agreement with in vitro experimental data. The model is used to simulate treatment by existing or potential new drugs, and to compare the efficacy of different schedules of drug delivery.
Collapse
Affiliation(s)
- Nourridine Siewe
- Department of Mathematics, Howard University, Washington, DC, United States.
| | - Abdul-Aziz Yakubu
- Department of Mathematics, Howard University, Washington, DC, United States
| | - Abhay R Satoskar
- Department of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, Ohio, United States
| | - Avner Friedman
- Department of Mathematics, Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
141
|
Yu YRA, O’Koren EG, Hotten DF, Kan MJ, Kopin D, Nelson ER, Que L, Gunn MD. A Protocol for the Comprehensive Flow Cytometric Analysis of Immune Cells in Normal and Inflamed Murine Non-Lymphoid Tissues. PLoS One 2016; 11:e0150606. [PMID: 26938654 PMCID: PMC4777539 DOI: 10.1371/journal.pone.0150606] [Citation(s) in RCA: 275] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/16/2016] [Indexed: 11/18/2022] Open
Abstract
Flow cytometry is used extensively to examine immune cells in non-lymphoid tissues. However, a method of flow cytometric analysis that is both comprehensive and widely applicable has not been described. We developed a protocol for the flow cytometric analysis of non-lymphoid tissues, including methods of tissue preparation, a 10-fluorochrome panel for cell staining, and a standardized gating strategy, that allows the simultaneous identification and quantification of all major immune cell types in a variety of normal and inflamed non-lymphoid tissues. We demonstrate that our basic protocol minimizes cell loss, reliably distinguishes macrophages from dendritic cells (DC), and identifies all major granulocytic and mononuclear phagocytic cell types. This protocol is able to accurately quantify 11 distinct immune cell types, including T cells, B cells, NK cells, neutrophils, eosinophils, inflammatory monocytes, resident monocytes, alveolar macrophages, resident/interstitial macrophages, CD11b- DC, and CD11b+ DC, in normal lung, heart, liver, kidney, intestine, skin, eyes, and mammary gland. We also characterized the expression patterns of several commonly used myeloid and macrophage markers. This basic protocol can be expanded to identify additional cell types such as mast cells, basophils, and plasmacytoid DC, or perform detailed phenotyping of specific cell types. In examining models of primary and metastatic mammary tumors, this protocol allowed the identification of several distinct tumor associated macrophage phenotypes, the appearance of which was highly specific to individual tumor cell lines. This protocol provides a valuable tool to examine immune cell repertoires and follow immune responses in a wide variety of tissues and experimental conditions.
Collapse
Affiliation(s)
- Yen-Rei A. Yu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| | - Emily G. O’Koren
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Danielle F. Hotten
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew J. Kan
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David Kopin
- School of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erik R. Nelson
- Department of Molecular & Integrative Physiology, University of Illinois, Champaign, Illinois, United States of America
| | - Loretta Que
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael D. Gunn
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
142
|
Khan ANH, Kolomeyevskaya N, Singel KL, Grimm MJ, Moysich KB, Daudi S, Grzankowski KS, Lele S, Ylagan L, Webster GA, Abrams SI, Odunsi K, Segal BH. Targeting myeloid cells in the tumor microenvironment enhances vaccine efficacy in murine epithelial ovarian cancer. Oncotarget 2016; 6:11310-26. [PMID: 25888637 PMCID: PMC4484458 DOI: 10.18632/oncotarget.3597] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/20/2015] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is typically diagnosed at advanced stages, and is associated with a high relapse rate. Patients in remission are ideal candidates for immunotherapy aimed at cure or prolonging disease-free periods. However, immunosuppressive pathways in the tumor microenvironment are obstacles to durable anti-tumor immunity. In a metastatic syngeneic mouse model of EOC, immunosuppressive macrophages and myeloid-derived suppressor cells (MDSCs) accumulate in the local tumor environment. In addition, resident peritoneal macrophages from non-tumor-bearing mice were highly immunosuppressive, abrogating stimulated T cell proliferation in a cell contact-dependent manner. Immunization with microparticles containing TLR9 and NOD-2 ligands (MIS416) significantly prolonged survival in tumor-bearing mice. The strategy of MIS416 immunization followed by anti-CD11b administration further delayed tumor progression, thereby establishing the proof of principle that myeloid depletion can enhance vaccine efficacy. In patients with advanced EOC, ascites analysis showed substantial heterogeneity in the relative proportions of myeloid subsets and their immunosuppressive properties. Together, these findings point to immunosuppressive myeloid cells in the EOC microenvironment as targets to enhance vaccination. Further studies of myeloid cell accumulation and functional phenotypes in the EOC microenvironment may identify patients who are likely to benefit from vaccination combined with approaches that deplete tumor-associated myeloid cells.
Collapse
Affiliation(s)
- Anm Nazmul H Khan
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Nonna Kolomeyevskaya
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kelly L Singel
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Melissa J Grimm
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sayeema Daudi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Sashikant Lele
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lourdes Ylagan
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Scott I Abrams
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Brahm H Segal
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Medicine, University at Buffalo School of Medicine, Buffalo, NY, USA
| |
Collapse
|
143
|
Daniotti JL, Lardone RD, Vilcaes AA. Dysregulated Expression of Glycolipids in Tumor Cells: From Negative Modulator of Anti-tumor Immunity to Promising Targets for Developing Therapeutic Agents. Front Oncol 2016; 5:300. [PMID: 26779443 PMCID: PMC4703717 DOI: 10.3389/fonc.2015.00300] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/14/2015] [Indexed: 12/14/2022] Open
Abstract
Glycolipids are complex molecules consisting of a ceramide lipid moiety linked to a glycan chain of variable length and structure. Among these are found the gangliosides, which are sialylated glycolipids ubiquitously distributed on the outer layer of vertebrate plasma membranes. Changes in the expression of certain species of gangliosides have been described to occur during cell proliferation, differentiation, and ontogenesis. However, the aberrant and elevated expression of gangliosides has been also observed in different types of cancer cells, thereby promoting tumor survival. Moreover, gangliosides are actively released from the membrane of tumor cells, having a strong impact on impairing anti-tumor immunity. Beyond the undesirable effects of gangliosides in cancer cells, a substantial number of cancer immunotherapies have been developed in recent years that have used gangliosides as the main target. This has resulted in successful immune cell- or antibody-responses against glycolipids, with promising results having been obtained in clinical trials. In this review, we provide a general overview on the metabolism of glycolipids, both in normal and tumor cells, as well as examining glycolipid-mediated immune modulation and the main successes achieved in immunotherapies using gangliosides as molecular targets.
Collapse
Affiliation(s)
- Jose Luis Daniotti
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , Córdoba , Argentina
| | - Ricardo D Lardone
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute at Providence Saint John's Health Center , Santa Monica, CA , USA
| | - Aldo A Vilcaes
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , Córdoba , Argentina
| |
Collapse
|
144
|
Chessa F, Mathow D, Wang S, Hielscher T, Atzberger A, Porubsky S, Gretz N, Burgdorf S, Gröne HJ, Popovic ZV. The renal microenvironment modifies dendritic cell phenotype. Kidney Int 2016; 89:82-94. [DOI: 10.1038/ki.2015.292] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/20/2015] [Accepted: 08/06/2015] [Indexed: 12/20/2022]
|
145
|
The Good, the Bad, and the Ugly of Dendritic Cells during Prion Disease. J Immunol Res 2015; 2015:168574. [PMID: 26697507 PMCID: PMC4677227 DOI: 10.1155/2015/168574] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/15/2015] [Indexed: 12/11/2022] Open
Abstract
Prions are a unique group of proteinaceous pathogens which cause neurodegenerative disease and can be transmitted by a variety of exposure routes. After peripheral exposure, the accumulation and replication of prions within secondary lymphoid organs are obligatory for their efficient spread from the periphery to the brain where they ultimately cause neurodegeneration and death. Mononuclear phagocytes (MNP) are a heterogeneous population of dendritic cells (DC) and macrophages. These cells are abundant throughout the body and display a diverse range of roles based on their anatomical locations. For example, some MNP are strategically situated to provide a first line of defence against pathogens by phagocytosing and destroying them. Conventional DC are potent antigen presenting cells and migrate via the lymphatics to the draining lymphoid tissue where they present the antigens to lymphocytes. The diverse roles of MNP are also reflected in various ways in which they interact with prions and in doing so impact on disease pathogenesis. Indeed, some studies suggest that prions exploit conventional DC to infect the host. Here we review our current understanding of the influence of MNP in the pathogenesis of the acquired prion diseases with particular emphasis on the role of conventional DC.
Collapse
|
146
|
Deng M, Ma T, Yan Z, Zettel KR, Scott MJ, Liao H, Frank A, Morelli AE, Sodhi CP, Hackam DJ, Billiar TR. Toll-like Receptor 4 Signaling on Dendritic Cells Suppresses Polymorphonuclear Leukocyte CXCR2 Expression and Trafficking via Interleukin 10 During Intra-abdominal Sepsis. J Infect Dis 2015; 213:1280-8. [PMID: 26603204 DOI: 10.1093/infdis/jiv562] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 11/09/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) is a critical receptor involved in the sensing of gram-negative bacterial infection. However, the roles of TLR4 in sepsis are cell-type specific. Dendritic cells (DCs) are known to play a central role in microbial detection, alerting the immune system to the presence of infection and coordinating adaptive immune response. The goal of this study was to investigate the impact of DC-specific TLR4 signaling on host defense against intra-abdominal polymicrobial sepsis. METHODS C57BL/6, global Tlr4 knockout, cell-specific knockout control, and CD11c-specific Tlr4(-/-) mice underwent cecal ligation and puncture (CLP). RESULTS Specific deletion of TLR4 on DCs in mice improved survival and enhanced bacterial clearance. Deletion of TLR4 on DCs was associated with lower levels of circulating interleukin 10 (IL-10), higher polymorphonuclear leukocyte (PMN) accumulation in the peritoneal cavity, and higher expression of chemokine (C-X-C motif) receptor 2 (CXCR2) on PMNs after CLP. In vitro studies of DC and neutrophil cocultures confirmed that TLR4-dependent secretion of IL-10 from DCs regulated neutrophil CXCR2 expression. CONCLUSIONS Our data shed light on a previously unrecognized role for TLR4 signaling on DCs in driving IL-10 secretion during sepsis and, through this pathway, regulates PMN recruitment via suppression of CXCR2 expression.
Collapse
Affiliation(s)
- Meihong Deng
- Department of Surgery, University of Pittsburgh, Pennsylvania
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital
| | - Zhengzheng Yan
- Department of Pathophysiology, Southern Medical University, Guangzhou, China
| | - Kent R Zettel
- Department of Surgery, University of Pittsburgh, Pennsylvania
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh, Pennsylvania
| | - Hong Liao
- Department of Surgery, University of Pittsburgh, Pennsylvania
| | - Alicia Frank
- Department of Surgery, University of Pittsburgh, Pennsylvania
| | | | | | | | | |
Collapse
|
147
|
Dendritic cells and macrophages neurally hard-wired in the lymph node. Sci Rep 2015; 5:16866. [PMID: 26581550 PMCID: PMC4652329 DOI: 10.1038/srep16866] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/21/2015] [Indexed: 12/25/2022] Open
Abstract
The neural hard-wired pathways in which the lymphoid organs are innervated by the nervous system is of special interest with respect to suggested afferent and sensory systems informing the central nervous system about the status of the immune system. Until today efferent also like afferent innervation seem to be unspecific, targeting many types of cells by affecting many cells at the same time. We for the first time show that antigen presenting cells (APC) are abundantly innervated in the T-cell enriched area, the subsinoidal layer and the cortical extrafollicular zone of lymph nodes in rats by a mesh of filamentous neurofilament positive structures originating from single nerve fibers and covering each single APC similar to a glass fishing float, so that we termed them “wired” APC (wAPC). These wAPC also found in humans seem to be restricted to the cell body, not to follow membranous extensions, they may be dynamic and receptive as MAP2 is expressed and axonal growth cones can be detected and they probably lack vesicular activity through missing synaptophysin expression. The specific innervation targeting single cells which show a distribution divided in several areas in one lymph node suggests a form of topographically organized afferent sensory system.
Collapse
|
148
|
Jung YJ, Jun HK, Choi BK. Gingipain-dependent augmentation by Porphyromonas gingivalis of phagocytosis of Tannerella forsythia. Mol Oral Microbiol 2015; 31:457-471. [PMID: 26434368 DOI: 10.1111/omi.12139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2015] [Indexed: 12/20/2022]
Abstract
In the pathogenesis of periodontitis, Porphyromonas gingivalis plays a role as a keystone pathogen that manipulates host immune responses leading to dysbiotic oral microbial communities. Arg-gingipains (RgpA and RgpB) and Lys-gingipain (Kgp) are responsible for the majority of bacterial proteolytic activity and play essential roles in bacterial virulence. Therefore, gingipains are often considered as therapeutic targets. This study investigated the role of gingipains in the modulation by P. gingivalis of phagocytosis of Tannerella forsythia by macrophages. Phagocytosis of T. forsythia was significantly enhanced by coinfection with P. gingivalis in a multiplicity of infection-dependent and gingipain-dependent manner. Mutation of either Kgp or Rgp in the coinfecting P. gingivalis resulted in attenuated enhancement of T. forsythia phagocytosis. Inhibition of coaggregation between the two bacterial species reduced phagocytosis of T. forsythia in mixed infection, and this coaggregation was dependent on gingipains. Inhibition of gingipain protease activities in coinfecting P. gingivalis abated the coaggregation and the enhancement of T. forsythia phagocytosis. However, the direct effect of protease activities of gingipains on T. forsythia seemed to be minimal. Although most of the phagocytosed T. forsythia were cleared in infected macrophages, more T. forsythia remained in cells coinfected with gingipain-expressing P. gingivalis than in cells coinfected with the gingipain-null mutant or infected only with T. forsythia at 24 and 48 h post-infection. Collectively, these results suggest that P. gingivalis, mainly via its gingipains, alters the clearance of T. forsythia, and provide some insights into the role of P. gingivalis as a keystone pathogen.
Collapse
Affiliation(s)
- Y-J Jung
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Korea
| | - H-K Jun
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Korea
| | - B-K Choi
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Korea. .,Dental Research Institute, Seoul National University, Seoul, Korea.
| |
Collapse
|
149
|
Kim M, Park SY, Jin ML, Park G, Son HJ. Cucurbitacin B inhibits immunomodulatory function and the inflammatory response in macrophages. Immunopharmacol Immunotoxicol 2015; 37:473-80. [DOI: 10.3109/08923973.2015.1085065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
150
|
Banat GA, Tretyn A, Pullamsetti SS, Wilhelm J, Weigert A, Olesch C, Ebel K, Stiewe T, Grimminger F, Seeger W, Fink L, Savai R. Immune and Inflammatory Cell Composition of Human Lung Cancer Stroma. PLoS One 2015; 10:e0139073. [PMID: 26413839 PMCID: PMC4587668 DOI: 10.1371/journal.pone.0139073] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 09/09/2015] [Indexed: 12/19/2022] Open
Abstract
Recent studies indicate that the abnormal microenvironment of tumors may play a critical role in carcinogenesis, including lung cancer. We comprehensively assessed the number of stromal cells, especially immune/inflammatory cells, in lung cancer and evaluated their infiltration in cancers of different stages, types and metastatic characteristics potential. Immunohistochemical analysis of lung cancer tissue arrays containing normal and lung cancer sections was performed. This analysis was combined with cyto-/histomorphological assessment and quantification of cells to classify/subclassify tumors accurately and to perform a high throughput analysis of stromal cell composition in different types of lung cancer. In human lung cancer sections we observed a significant elevation/infiltration of total-T lymphocytes (CD3+), cytotoxic-T cells (CD8+), T-helper cells (CD4+), B cells (CD20+), macrophages (CD68+), mast cells (CD117+), mononuclear cells (CD11c+), plasma cells, activated-T cells (MUM1+), B cells, myeloid cells (PD1+) and neutrophilic granulocytes (myeloperoxidase+) compared with healthy donor specimens. We observed all of these immune cell markers in different types of lung cancers including squamous cell carcinoma, adenocarcinoma, adenosquamous cell carcinoma, small cell carcinoma, papillary adenocarcinoma, metastatic adenocarcinoma, and bronchioloalveolar carcinoma. The numbers of all tumor-associated immune cells (except MUM1+ cells) in stage III cancer specimens was significantly greater than those in stage I samples. We observed substantial stage-dependent immune cell infiltration in human lung tumors suggesting that the tumor microenvironment plays a critical role during lung carcinogenesis. Strategies for therapeutic interference with lung cancer microenvironment should consider the complexity of its immune cell composition.
Collapse
Affiliation(s)
- G-Andre Banat
- Internal Medicine, University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Aleksandra Tretyn
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
| | - Soni Savai Pullamsetti
- Internal Medicine, University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
| | - Jochen Wilhelm
- Internal Medicine, University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Catherine Olesch
- Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Katharina Ebel
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
| | - Thorsten Stiewe
- Molecular Oncology, Philipps-University, Member of the German Center for Lung Research, Marburg, Germany
| | - Friedrich Grimminger
- Internal Medicine, University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Werner Seeger
- Internal Medicine, University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
| | - Ludger Fink
- Institute of Pathology and Cytology, UEGP, Wetzlar, Germany
| | - Rajkumar Savai
- Internal Medicine, University of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
- * E-mail:
| |
Collapse
|