Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Immunity ; 51(4): 638-654.e9, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31561945

ABSTRACT

Macrophages are strongly adapted to their tissue of residence. Yet, little is known about the cell-cell interactions that imprint the tissue-specific identities of macrophages in their respective niches. Using conditional depletion of liver Kupffer cells, we traced the developmental stages of monocytes differentiating into Kupffer cells and mapped the cellular interactions imprinting the Kupffer cell identity. Kupffer cell loss induced tumor necrosis factor (TNF)- and interleukin-1 (IL-1) receptor-dependent activation of stellate cells and endothelial cells, resulting in the transient production of chemokines and adhesion molecules orchestrating monocyte engraftment. Engrafted circulating monocytes transmigrated into the perisinusoidal space and acquired the liver-associated transcription factors inhibitor of DNA 3 (ID3) and liver X receptor-α (LXR-α). Coordinated interactions with hepatocytes induced ID3 expression, whereas endothelial cells and stellate cells induced LXR-α via a synergistic NOTCH-BMP pathway. This study shows that the Kupffer cell niche is composed of stellate cells, hepatocytes, and endothelial cells that together imprint the liver-specific macrophage identity.


Subject(s)
Endothelial Cells/physiology , Hepatic Stellate Cells/physiology , Hepatocytes/physiology , Kupffer Cells/physiology , Liver/cytology , Macrophages/physiology , Monocytes/physiology , Animals , Cell Communication , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Female , Gene Expression Regulation , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Notch/metabolism
2.
Immunity ; 49(2): 312-325.e5, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30076102

ABSTRACT

Heterogeneity between different macrophage populations has become a defining feature of this lineage. However, the conserved factors defining macrophages remain largely unknown. The transcription factor ZEB2 is best described for its role in epithelial to mesenchymal transition; however, its role within the immune system is only now being elucidated. We show here that Zeb2 expression is a conserved feature of macrophages. Using Clec4f-cre, Itgax-cre, and Fcgr1-cre mice to target five different macrophage populations, we found that loss of ZEB2 resulted in macrophage disappearance from the tissues, coupled with their subsequent replenishment from bone-marrow precursors in open niches. Mechanistically, we found that ZEB2 functioned to maintain the tissue-specific identities of macrophages. In Kupffer cells, ZEB2 achieved this by regulating expression of the transcription factor LXRα, removal of which recapitulated the loss of Kupffer cell identity and disappearance. Thus, ZEB2 expression is required in macrophages to preserve their tissue-specific identities.


Subject(s)
Kupffer Cells/cytology , Liver X Receptors/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Lineage/immunology , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Kupffer Cells/immunology , Liver/cytology , Liver X Receptors/metabolism , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Am J Pathol ; 193(4): 366-379, 2023 04.
Article in English | MEDLINE | ID: mdl-36642171

ABSTRACT

Primary sclerosing cholangitis (PSC) is an idiopathic chronic immune-mediated cholestatic liver disease characterized by fibro-inflammatory bile duct strictures, progressive hepatobiliary fibrosis, and gut-liver axis disruption. The pathophysiology of PSC remains insufficiently characterized, which hampers the development of effective therapies. Hepatic macrophages (MFs) such as Kupffer cells (KCs) are implicated in PSC pathogenesis, but their exact role is unclear. Using the latest markers to discriminate resident KCs (ResKCs) from their monocyte-derived counterparts (MoKCs), and two models of intrahepatic and extrahepatic cholestasis, respectively, this study showed that CLEC4F+TIM4+ ResKCs were depleted after chronic cholestatic liver injury. The infiltrating CLEC4F+TIM4- MoKCs were already enriched during the acute phase of PSC. Transcriptional profiling of hepatic MF subsets during early cholestatic injury indicated that ResKCs were indeed activated and that MoKCs expressed higher levels of pro-inflammatory and proliferative markers compared with those of ResKCs. As indicated in experiments with Clec4fDTR transgenic mice, conditional depletion of KCs, before and during early cholestasis induction, had no effect on the composition of the hepatic myeloid cell pool following injury progression and did not affect disease outcomes. Taken together, these results provide new insights into the heterogeneity of the MF pool during experimental PSC and evidence that depletion of resident and activated KCs during sclerosing cholangitis does not affect disease outcome in mice.


Subject(s)
Cholangitis, Sclerosing , Cholestasis , Mice , Animals , Cholangitis, Sclerosing/pathology , Kupffer Cells/pathology , Liver/pathology , Cholestasis/pathology
4.
Int J Cancer ; 152(12): 2615-2628, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36912275

ABSTRACT

Due to a combination of rapid disease progression and the lack of curative treatment options, hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Infiltrated, monocyte-derived, tumor-associated macrophages are known to play a role in HCC pathogenesis, but the involvement of Kupffer cells (KCs) remains elusive. Here, we used the Clec4F-diphteria toxin receptor transgenic mouse model to specifically investigate the effect of KC depletion on HCC initiation, progression and neoplastic growth following liver resection. For this purpose, several HCC mouse models with varying underlying etiologies were used and partial hepatectomy was performed. Our results show that in HCC, developed on a fibrotic or non-alcoholic steatohepatitis background, depletion of embryonic KCs at the onset of HCC induction and the subsequent replacement by monocyte-derived KCs does not affect the tumor burden, tumor microenvironment or the phenotype of isolated KCs at end-stage disease. In non-chronic liver disease-associated diethylnitrosamine-induced HCC, ablation of Clec4F+ KCs did not alter tumor progression or neoplastic growth following liver resection. Our results show that temporal ablation of resident KCs does not impact HCC pathogenesis, neither in the induction phase nor in advanced disease, and indicate that bone marrow-derived KCs are able to swiftly repopulate the available KC niche and adopt their phenotype.


Subject(s)
Carcinogenesis , Carcinoma, Hepatocellular , Kupffer Cells , Liver Neoplasms, Experimental , Liver Neoplasms , Tumor-Associated Macrophages , Kupffer Cells/immunology , Disease Progression , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Animals , Mice , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Monocyte-Macrophage Precursor Cells/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Mice, Inbred C57BL , Male
5.
J Immunol ; 199(5): 1762-1771, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28739879

ABSTRACT

Arginase activity induction in macrophages is an escape mechanism developed by parasites to cope with the host's immune defense and benefit from increased host-derived growth factor production. We report that arginase expression and activity were induced in macrophages during mouse infection by Trypanosoma musculi, a natural parasite of this host. This induction was reproduced in vitro by excreted/secreted factors of the parasite. A mAb directed to TbKHC1, an orphan kinesin H chain from Trypanosoma brucei, inhibited T. musculi excreted/secreted factor-mediated arginase induction. Anti-TbKHC1 Ab also inhibited T. musculi growth, both in vitro and in vivo. Induction of arginase activity and parasite growth involved C-type lectin receptors, because mannose injection decreased arginase activity induction and parasite load in vitro and in vivo. Accordingly, the parasite load was reduced in mice lacking mannose receptor C-type 1. The T. musculi KHC1 homolog showed high similarity with TbKHC1. Bioinformatics analysis revealed the presence of homologs of this gene in other trypanosomes, including pathogens for humans and animals. Host metabolism dysregulation represents an effective parasite mechanism to hamper the host immune response and modify host molecule production to favor parasite invasion and growth. Thus, this orphan kinesin plays an important role in promoting trypanosome infection, and its neutralization or the lock of its partner host molecules offers promising approaches to increasing resistance to infection and new developments in vaccination against trypanosomiasis.


Subject(s)
Antigens, Protozoan/metabolism , Arginase/metabolism , Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Macrophages/immunology , Receptors, Cell Surface/metabolism , Trypanosoma/physiology , Trypanosomiasis/immunology , Animals , Antibodies/metabolism , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Cell Adhesion Molecules/genetics , Cells, Cultured , Female , Kinesins/genetics , Lectins, C-Type/genetics , Macrophages/parasitology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Parasite Load , Phylogeny , Receptors, Cell Surface/genetics , Vaccination
6.
PLoS Pathog ; 12(9): e1005862, 2016 09.
Article in English | MEDLINE | ID: mdl-27632207

ABSTRACT

Animal African trypanosomosis is a major threat to the economic development and human health in sub-Saharan Africa. Trypanosoma congolense infections represent the major constraint in livestock production, with anemia as the major pathogenic lethal feature. The mechanisms underlying anemia development are ill defined, which hampers the development of an effective therapy. Here, the contribution of the erythropoietic and erythrophagocytic potential as well as of hemodilution to the development of T. congolense-induced anemia were addressed in a mouse model of low virulence relevant for bovine trypanosomosis. We show that in infected mice, splenic extramedullary erythropoiesis could compensate for the chronic low-grade type I inflammation-induced phagocytosis of senescent red blood cells (RBCs) in spleen and liver myeloid cells, as well as for the impaired maturation of RBCs occurring in the bone marrow and spleen. Rather, anemia resulted from hemodilution. Our data also suggest that the heme catabolism subsequent to sustained erythrophagocytosis resulted in iron accumulation in tissue and hyperbilirubinemia. Moreover, hypoalbuminemia, potentially resulting from hemodilution and liver injury in infected mice, impaired the elimination of toxic circulating molecules like bilirubin. Hemodilutional thrombocytopenia also coincided with impaired coagulation. Combined, these effects could elicit multiple organ failure and uncontrolled bleeding thus reduce the survival of infected mice. MIF (macrophage migrating inhibitory factor), a potential pathogenic molecule in African trypanosomosis, was found herein to promote erythrophagocytosis, to block extramedullary erythropoiesis and RBC maturation, and to trigger hemodilution. Hence, these data prompt considering MIF as a potential target for treatment of natural bovine trypanosomosis.


Subject(s)
Anemia/metabolism , Erythropoiesis , Hematopoiesis, Extramedullary , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Trypanosoma congolense/metabolism , Trypanosomiasis, African/metabolism , Anemia/genetics , Anemia/parasitology , Anemia/pathology , Animals , Bone Marrow/metabolism , Bone Marrow/parasitology , Bone Marrow/pathology , Cattle , Disease Models, Animal , Erythrocytes/metabolism , Erythrocytes/parasitology , Erythrocytes/pathology , Hemodilution , Humans , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Knockout , Spleen/metabolism , Spleen/parasitology , Spleen/pathology , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/parasitology , Thrombocytopenia/pathology , Trypanosomiasis, African/genetics , Trypanosomiasis, African/pathology
7.
Cell Immunol ; 330: 202-215, 2018 08.
Article in English | MEDLINE | ID: mdl-29661474

ABSTRACT

The liver is a major target organ for metastasis of both gastrointestinal and extra-gastrointestinal cancers. Due to its frequently inoperable nature, liver metastasis represents a leading cause of cancer-associated death worldwide. In the past years, the pivotal role of the immune system in this process is being increasingly recognised. In particular, the role of the hepatic macrophages, both recruited monocyte-derived macrophages (Mo-Mfs) and tissue-resident Kupffer cells (KCs), has been shown to be more versatile than initially imagined. However, the lack of tools to easily distinguish between these two macrophage populations has hampered the assignment of particular functionalities to specific hepatic macrophage subsets. In this Review, we highlight the most remarkable findings regarding the origin and functions of hepatic macrophage populations, and we provide a detailed description of their distinct roles in the different phases of the liver metastatic process.


Subject(s)
Kupffer Cells/immunology , Liver Neoplasms/immunology , Liver/immunology , Macrophages/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Hepatocytes/immunology , Hepatocytes/metabolism , Homeostasis/immunology , Humans , Kupffer Cells/pathology , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Metastasis
8.
Biochim Biophys Acta ; 1862(6): 1111-21, 2016 06.
Article in English | MEDLINE | ID: mdl-26912412

ABSTRACT

BACKGROUND AND AIMS: Being goalkeepers of liver homeostasis, gap junctions are also involved in hepatotoxicity. However, their role in this process is ambiguous, as gap junctions can act as both targets and effectors of liver toxicity. This particularly holds true for drug-induced liver insults. In the present study, the involvement of connexin26, connexin32 and connexin43, the building blocks of liver gap junctions, was investigated in acetaminophen-induced hepatotoxicity. METHODS: C57BL/6 mice were overdosed with 300mg/kg body weight acetaminophen followed by analysis of the expression and localization of connexins as well as monitoring of hepatic gap junction functionality. Furthermore, acetaminophen-induced liver injury was compared between mice genetically deficient in connexin43 and wild type littermates. Evaluation of the toxicological response was based on a set of clinically relevant parameters, including protein adduct formation, measurement of alanine aminotransferase activity, cytokines and glutathione. RESULTS: It was found that gap junction communication deteriorates upon acetaminophen intoxication in wild type mice, which is associated with a switch in mRNA and protein production from connexin32 and connexin26 to connexin43. The upregulation of connexin43 expression is due, at least in part, to de novo production by hepatocytes. Connexin43-deficient animals tended to show increased liver cell death, inflammation and oxidative stress in comparison with wild type counterparts. CONCLUSION: These results suggest that hepatic connexin43-based signaling may protect against acetaminophen-induced liver toxicity.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Chemical and Drug Induced Liver Injury/genetics , Connexin 43/genetics , Liver/drug effects , Up-Regulation/drug effects , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Connexin 43/analysis , Connexin 43/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , RNA, Messenger/analysis , RNA, Messenger/genetics
9.
PLoS Pathog ; 11(5): e1004873, 2015 May.
Article in English | MEDLINE | ID: mdl-26020782

ABSTRACT

Monocytes consist of two well-defined subsets, the Ly6C+ and Ly6C- monocytes. Both CD11b+ myeloid cells populations have been proposed to infiltrate tissues during inflammation. While infiltration of Ly6C+ monocytes is an established pathogenic factor during hepatic inflammation, the role of Ly6C- monocytes remains elusive. Mice suffering experimental African trypanosome infection die from systemic inflammatory response syndrome (SIRS) that is initiated by phagocytosis of parasites by liver myeloid cells and culminates in apoptosis/necrosis of liver myeloid and parenchymal cells that reduces host survival. C57BL/6 mice are considered as trypanotolerant to Trypanosoma congolense infection. We have reported that in these animals, IL-10, produced among others by myeloid cells, limits the liver damage caused by pathogenic TNF-producing Ly6C+ monocytes, ensuring prolonged survival. Here, the heterogeneity and dynamics of liver myeloid cells in T. congolense-infected C57/BL6 mice was further dissected. Moreover, the contribution of Ly6C- monocytes to trypanotolerance was investigated. By using FACS analysis and adoptive transfer experiments, we found that the accumulation of Ly6C- monocytes and macrophages in the liver of infected mice coincided with a drop in the pool of Ly6C+ monocytes. Pathogenic TNF mainly originated from Ly6C+ monocytes while Ly6C- monocytes and macrophages were major and equipotent sources of IL-10 within myeloid cells. Moreover, Nr4a1 (Nur77) transcription factor-dependent Ly6C- monocytes exhibited IL-10-dependent and cell contact-dependent regulatory properties contributing to trypanotolerance by suppressing the production of TNF by Ly6C+ monocytes and by promoting the differentiation of the latter cells into macrophages. Thus, Ly6C- monocytes can dampen liver damage caused by an extensive Ly6C+ monocyte-associated inflammatory immune response in T. congolense trypanotolerant animals. In a more general context, Ly6C- or Ly6C+ monocyte targeting may represent a therapeutic approach in liver pathogenicity induced by chronic infection.


Subject(s)
Antigens, Ly/immunology , Cell Differentiation , Inflammation/etiology , Liver Diseases/etiology , Macrophages/immunology , Monocytes/immunology , Monocytes/pathology , Trypanosomiasis, African/immunology , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Female , Flow Cytometry , Immunoenzyme Techniques , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/metabolism , Liver Diseases/pathology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/pathology , Phagocytosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Trypanosoma congolense/immunology , Trypanosomiasis, African/complications , Trypanosomiasis, African/parasitology , Tumor Cells, Cultured
10.
Arch Toxicol ; 91(5): 2245-2261, 2017 May.
Article in English | MEDLINE | ID: mdl-27826632

ABSTRACT

Pannexins constitute a relatively new family of transmembrane proteins that form channels linking the cytoplasmic compartment with the extracellular environment. The presence of pannexin1 in the liver has been documented previously, where it underlies inflammatory responses, such as those occurring upon ischemia-reperfusion injury. In the present study, we investigated whether pannexin1 plays a role in acute drug-induced liver toxicity. Hepatic expression of pannexin1 was characterized in a mouse model of acetaminophen-induced hepatotoxicity. Subsequently, mice were overdosed with acetaminophen followed by treatment with the pannexin1 channel inhibitor 10Panx1. Sampling was performed 1, 3, 6, 24 and 48 h after acetaminophen administration. Evaluation of the effects of pannexin1 channel inhibition was based on a number of clinically relevant readouts, including protein adduct formation, measurement of aminotransferase activity and histopathological examination of liver tissue as well as on a series of markers of inflammation, oxidative stress and regeneration. Although no significant differences were found in histopathological analysis, pannexin1 channel inhibition reduced serum levels of alanine and aspartate aminotransferase. This was paralleled by a reduced amount of neutrophils recruited to the liver. Furthermore, alterations in the oxidized status were noticed with upregulation of glutathione levels upon suppression of pannexin1 channel opening. Concomitant promotion of regenerative activity was detected as judged on increased proliferating cell nuclear antigen protein quantities in 10Panx1-treated mice. Pannexin1 channels are important actors in liver injury triggered by acetaminophen. Inhibition of pannexin1 channel opening could represent a novel approach for the treatment of drug-induced hepatotoxicity.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/drug therapy , Connexins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Connexins/genetics , Connexins/metabolism , Cytokines/blood , Cytokines/metabolism , Drug Overdose/metabolism , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neutrophils/drug effects , Oxidative Stress/drug effects
11.
PLoS Pathog ; 10(9): e1004414, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25255103

ABSTRACT

African trypanosomiasis is a chronic debilitating disease affecting the health and economic well-being of many people in developing countries. The pathogenicity associated with this disease involves a persistent inflammatory response, whereby M1-type myeloid cells, including Ly6C(high) inflammatory monocytes, are centrally implicated. A comparative gene analysis between trypanosusceptible and trypanotolerant animals identified MIF (macrophage migrating inhibitory factor) as an important pathogenic candidate molecule. Using MIF-deficient mice and anti-MIF antibody treated mice, we show that MIF mediates the pathogenic inflammatory immune response and increases the recruitment of inflammatory monocytes and neutrophils to contribute to liver injury in Trypanosoma brucei infected mice. Moreover, neutrophil-derived MIF contributed more significantly than monocyte-derived MIF to increased pathogenic liver TNF production and liver injury during trypanosome infection. MIF deficient animals also featured limited anemia, coinciding with increased iron bio-availability, improved erythropoiesis and reduced RBC clearance during the chronic phase of infection. Our data suggest that MIF promotes the most prominent pathological features of experimental trypanosome infections (i.e. anemia and liver injury), and prompt considering MIF as a novel target for treatment of trypanosomiasis-associated immunopathogenicity.


Subject(s)
Anemia/immunology , Apoptosis/immunology , Erythrocytes/immunology , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Macrophages/immunology , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/immunology , Anemia/metabolism , Anemia/parasitology , Anemia/pathology , Animals , Blotting, Western , Bone Marrow/immunology , Bone Marrow/parasitology , Bone Marrow/pathology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Erythrocytes/pathology , Female , Flow Cytometry , Liver/immunology , Liver/parasitology , Liver/pathology , Macrophages/metabolism , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/parasitology , Monocytes/pathology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/parasitology , Neutrophils/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Spleen/metabolism , Spleen/parasitology , Spleen/pathology , Trypanosomiasis, African/metabolism , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/pathology
12.
PLoS Pathog ; 9(10): e1003731, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204274

ABSTRACT

BACKGROUND: In order to promote infection, the blood-borne parasite Trypanosoma brucei releases factors that upregulate arginase expression and activity in myeloid cells. METHODOLOGY/PRINCIPAL FINDINGS: By screening a cDNA library of T. brucei with an antibody neutralizing the arginase-inducing activity of parasite released factors, we identified a Kinesin Heavy Chain isoform, termed TbKHC1, as responsible for this effect. Following interaction with mouse myeloid cells, natural or recombinant TbKHC1 triggered SIGN-R1 receptor-dependent induction of IL-10 production, resulting in arginase-1 activation concomitant with reduction of nitric oxide (NO) synthase activity. This TbKHC1 activity was IL-4Rα-independent and did not mirror M2 activation of myeloid cells. As compared to wild-type T. brucei, infection by TbKHC1 KO parasites was characterized by strongly reduced parasitaemia and prolonged host survival time. By treating infected mice with ornithine or with NO synthase inhibitor, we observed that during the first wave of parasitaemia the parasite growth-promoting effect of TbKHC1-mediated arginase activation resulted more from increased polyamine production than from reduction of NO synthesis. In late stage infection, TbKHC1-mediated reduction of NO synthesis appeared to contribute to liver damage linked to shortening of host survival time. CONCLUSION: A kinesin heavy chain released by T. brucei induces IL-10 and arginase-1 through SIGN-R1 signaling in myeloid cells, which promotes early trypanosome growth and favors parasite settlement in the host. Moreover, in the late stage of infection, the inhibition of NO synthesis by TbKHC1 contributes to liver pathogenicity.


Subject(s)
Arginase/immunology , Kinesins/immunology , Protozoan Proteins/immunology , Trypanosoma brucei brucei/immunology , Trypanosomiasis, African/immunology , Animals , Arginase/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Kinesins/genetics , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide/immunology , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/genetics , Trypanosomiasis, African/pathology
13.
J Pathol ; 229(2): 186-97, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23011901

ABSTRACT

Accumulation of extracellular matrix components secreted by fibroblasts is a normal feature of wound healing during acute inflammation. However, during most chronic/persistent inflammatory diseases, this tissue repair mechanism is incorrectly regulated and results in irreversible fibrosis in various organs. Fibrosis that severely affects tissue architecture and can cause organ failure is a major cause of death in developed countries. Organ-recruited lymphoid (mainly T cells) and myeloid cells (eosinophils, basophils, macrophages and DCs) have long been recognized in their participation to the development of fibrosis. In particular, a central role for recruited monocyte-derived macrophages in this excessive connective tissue deposit is more and more appreciated. Moreover, the polarization of monocyte-derived macrophages in classically activated (IFNγ-dependent) M1 cells or alternatively activated (IL-4/IL-13) M2 cells, that mirrors the Th1/Th2 polarization of T cells, is also documented to contribute differentially to the fibrotic process. Here, we review the current understanding of how myeloid cell subpopulations affect the development of fibrosis in parasite infections.


Subject(s)
Liver Cirrhosis/parasitology , Liver Diseases, Parasitic/parasitology , Liver/parasitology , Myeloid Cells/parasitology , Animals , Echinococcosis, Hepatic/immunology , Echinococcosis, Hepatic/parasitology , Echinococcosis, Hepatic/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/parasitology , Extracellular Matrix/pathology , Humans , Inflammation Mediators/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Diseases, Parasitic/immunology , Liver Diseases, Parasitic/metabolism , Liver Diseases, Parasitic/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Schistosomiasis/immunology , Schistosomiasis/parasitology , Schistosomiasis/pathology
14.
Cell Rep ; 43(2): 113757, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38354088

ABSTRACT

Understanding the mechanisms underlying cytotoxic immunoglobulin G (IgG) activity is critical for improving therapeutic antibody activity and inhibiting autoantibody-mediated tissue pathology. While prior research highlights the important role of the mononuclear phagocytic system for removing opsonized target cells, it remains unclear which monocyte or macrophage subsets stemming from fetal or post-natal bone-marrow (BM)-associated definitive hematopoiesis are involved in target cell depletion. By using a titrated irradiation approach as well as Kupffer-cell-specific deletion of activated Fcγ receptor signaling, we establish conditions under which the contribution of BM-derived monocytes versus yolk-sac-derived liver-resident macrophages to cytotoxic IgG activity can be studied. Our results demonstrate that liver-resident macrophages originating from either fetal or adult hematopoiesis play a central role in IgG-mediated depletion of opsonized target cells from the peripheral blood under steady-state conditions, highlighting the impact of the tissue niche and not macrophage origin for cytotoxic antibody activity.


Subject(s)
Bone Marrow , Immunoglobulin G , Adult , Humans , Fetus , Macrophages , Monocytes
15.
Eur J Immunol ; 42(11): 2971-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22806454

ABSTRACT

Recently, we identified the CD20 homolog Ms4a8a as a novel molecule expressed by tumor-associated macrophages that directly enhances tumor growth. Here, we analyzed Ms4a8a(+) macrophages in M2-associated infectious pathologies. In late-stage Trypanosoma congolense and Taenia crassiceps infections, Ms4a8a expression was detected in hepatic and peritoneal macrophages respectively. Innate immunity in these infections is modulated by Toll-like receptor (TLR) signaling and TLR2/4/7 agonists strongly induced Ms4a8a expression in bone marrow derived macrophages (BMDMs) treated with M2 mediators (glucocorticoids/IL-4). LPS/dexamethasone/IL-4-induced Ms4a8a(+) BMDMs were characterized by strong expression of mRNA of mannose receptor (Mmr), arginase 1, and CD163, and by decreased iNOS expression. Coinduction of Ms4a8a by M2 mediators and TLR agonists involved the classical TLR signaling cascade via activation of MyD88/TRIF and NF-κB. Forced overexpression of Ms4a8a modulated the TLR4 response of RAW264.7 cells as shown by gene expression profiling. Upregulation of Hdc, Tcfec, and Sla was confirmed both in primary LPS/dexamethasone/IL-4-stimulated Ms4a8a(+) BMDMs and in peritoneal macrophages from late-stage Taenia crassiceps infection. In conclusion, we show that TLR signaling skews the typical alternative macrophage activation program to induce a special M2-like macrophage subset in vitro that also occurs in immunomodulatory immune reactions in vivo, a process directly involving the CD20 homolog Ms4a8a.


Subject(s)
Antigens, CD20/immunology , Macrophages/immunology , Taenia/immunology , Taeniasis/immunology , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , Arginase/genetics , Arginase/immunology , Cell Line , Immunity, Innate/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Macrophage Activation/immunology , Macrophages/parasitology , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA/chemistry , RNA/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Specific Pathogen-Free Organisms , Taeniasis/parasitology , Toll-Like Receptors/agonists , Trypanosomiasis, African/parasitology
16.
J Immunol ; 187(5): 2656-65, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21804013

ABSTRACT

Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects several million people in Latin America. Myocarditis, observed in the acute and chronic phases of the disease, is characterized by a mononuclear cell inflammatory infiltrate. We previously identified a myeloid cell population in the inflammatory heart infiltrate of infected mice that expressed arginase I. In this study, we purified CD11b(+) myeloid cells from the heart and analyzed their phenotype and function. Those CD11b(+) cells were ∼70% Ly6G(-)Ly6C(+) and 25% Ly6G(+)Ly6C(+). Moreover, purified CD11b(+)Ly6G(-) cells, but not Ly6G(+) cells, showed a predominant monocytic phenotype, expressed arginase I and inducible NO synthase, and suppressed anti-CD3/anti-CD28 Ab-induced T cell proliferation in vitro by an NO-dependent mechanism, activity that best defines myeloid-derived suppressor cells (MDSCs). Contrarily, CD11b(+)Ly6G(+) cells, but not CD11b(+)Ly6G(-) cells, expressed S100A8 and S100A9, proteins known to promote recruitment and differentiation of MDSCs. Together, our results suggest that inducible NO synthase/arginase I-expressing CD11b(+)Ly6G(-) myeloid cells in the hearts of T. cruzi-infected mice are MDSCs. Finally, we found plasma l-arginine depletion in the acute phase of infection that was coincident in time with the appearance of MDSCs, suggesting that in vivo arginase I could be contributing to l-arginine depletion and systemic immunosuppression. Notably, l-arginine supplementation decreased heart tissue parasite load, suggesting that sustained arginase expression through the acute infection is detrimental for the host. This is, to our knowledge, the first time that MDSCs have been found in the heart in the context of myocarditis and also in infection by T. cruzi.


Subject(s)
Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Myeloid Cells/metabolism , Animals , Arginase/metabolism , Arginine/blood , CD11b Antigen/biosynthesis , Cell Separation , Chagas Cardiomyopathy/immunology , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/immunology , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Trypanosoma cruzi/immunology
17.
Eur J Immunol ; 41(12): 3479-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21928284

ABSTRACT

DCs represent the major cell type leading to polarized T-helper (Th) cell responses in vivo. Here, we asked whether the instruction of murine Th2 responses by DCs matured with the proinflammatory cytokine TNF is qualitatively different from maturation by different types of TLR4/MyD88-dependent variant-specific surface glycoproteins (VSGs) of Trypanosoma brucei (T. brucei). The results obtained by analyzing DC surface markers, Notch ligand mRNA, cytokines, asthma, and experimental autoimmune encephalomyelitis (EAE) models as well as performing microarrays indicate that both types of stimuli induce similar inflammatory, semi-mature DC profiles. DCs matured by TNF or VSG treatment expressed a common inflammatory signature of 24 genes correlating with their Th2-polarization capacity. However, the same 24 genes and 4498 additional genes were expressed by DCs treated with LPS that went on to induce Th1 cells. These findings support the concept of a default pathway for Th2-cell induction in DCs matured under suboptimal or inflammatory conditions, independent of the surface receptors and signaling pathways involved. Our data also indicate that quantitative differences in DC maturation might direct Th2- vs Th1-cell responses, since suboptimally matured inflammatory DCs induce default Th2-cell maturation, whereas fully mature DCs induce Th1-cell maturation.


Subject(s)
Antigens, Protozoan/immunology , Dendritic Cells/immunology , Inflammation/immunology , Th2 Cells/immunology , Trypanosoma brucei brucei/immunology , Tumor Necrosis Factor-alpha/immunology , Variant Surface Glycoproteins, Trypanosoma/immunology , Animals , Asthma/immunology , Asthma/metabolism , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Profiling/methods , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/immunology , Th1 Cells/immunology , Up-Regulation/immunology , Variant Surface Glycoproteins, Trypanosoma/metabolism
18.
Eur J Immunol ; 41(11): 3270-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21805465

ABSTRACT

A balance between parasite elimination and control of infection-associated pathogenicity is crucial for resistance to African trypanosomiasis. By producing TNF and NO, CD11b(+) myeloid cells with a classical activation status (M1) contribute to parasitemia control in experimental Trypanosoma congolense infection in resistant C57BL/6 mice. However, in these mice, IL-10 is required to regulate M1-associated inflammation, avoiding tissue/liver damage and ensuring prolonged survival. In an effort to dissect the mechanisms behind the anti-inflammatory activity of IL-10 in T. congolense-infected C57BL/6 mice, we show, using an antibody blocking the IL-10 receptor, that IL-10 impairs the accumulation and M1 activation of TNF/iNOS-producing CD11b(+) Ly6C(+) cells in the liver. Using infected IL-10(flox/flox) LysM-Cre(+/+) mice, we show that myeloid cell-derived IL-10 limits M1 activation of CD11b(+) Ly6C(+) cells specifically at the level of TNF production. Moreover, higher production of TNF in infected IL-10(flox/flox) LysM-Cre(+/+) mice is associated with reduced nuclear accumulation of the NF-κB p50 subunit in CD11b(+) M1 cells. Furthermore, in infected p50(-/-) mice, TNF production by CD11b(+) Ly6C(+) cells and liver injury increases. These data suggest that preferential nuclear accumulation of p50 represents an IL-10-dependent anti-inflammatory mechanism in M1-type CD11b(+) myeloid cells that regulates the production of pathogenic TNF during T. congolense infection in resistant C57BL/6 mice.


Subject(s)
Interleukin-10/immunology , Myeloid Cells/immunology , NF-kappa B p50 Subunit/immunology , Trypanosoma congolense/immunology , Trypanosomiasis, African/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Blotting, Western , Cell Separation , Flow Cytometry , Interleukin-10/metabolism , Liver/cytology , Liver/immunology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , NF-kappa B p50 Subunit/metabolism , Signal Transduction/immunology , Trypanosomiasis, African/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
19.
PLoS Pathog ; 6(8): e1001045, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20714353

ABSTRACT

The development of classically activated monocytic cells (M1) is a prerequisite for effective elimination of parasites, including African trypanosomes. However, persistent activation of M1 that produce pathogenic molecules such as TNF and NO contributes to the development of trypanosome infection-associated tissue injury including liver cell necrosis in experimental mouse models. Aiming to identify mechanisms involved in regulation of M1 activity, we have recently documented that during Trypanosoma brucei infection, CD11b(+)Ly6C(+)CD11c(+) TNF and iNOS producing DCs (Tip-DCs) represent the major pathogenic M1 liver subpopulation. By using gene expression analyses, KO mice and cytokine neutralizing antibodies, we show here that the conversion of CD11b(+)Ly6C(+) monocytic cells to pathogenic Tip-DCs in the liver of T. brucei infected mice consists of a three-step process including (i) a CCR2-dependent but CCR5- and Mif-independent step crucial for emigration of CD11b(+)Ly6C(+) monocytic cells from the bone marrow but dispensable for their blood to liver migration; (ii) a differentiation step of liver CD11b(+)Ly6C(+) monocytic cells to immature inflammatory DCs (CD11c(+) but CD80/CD86/MHC-II(low)) which is IFN-gamma and MyD88 signaling independent; and (iii) a maturation step of inflammatory DCs to functional (CD80/CD86/MHC-II(high)) TNF and NO producing Tip-DCs which is IFN-gamma and MyD88 signaling dependent. Moreover, IL-10 could limit CCR2-mediated egression of CD11b(+)Ly6C(+) monocytic cells from the bone marrow by limiting Ccl2 expression by liver monocytic cells, as well as their differentiation and maturation to Tip-DCs in the liver, showing that IL-10 works at multiple levels to dampen Tip-DC mediated pathogenicity during T. brucei infection. A wide spectrum of liver diseases associates with alteration of monocyte recruitment, phenotype or function, which could be modulated by IL-10. Therefore, investigating the contribution of recruited monocytes to African trypanosome induced liver injury could potentially identify new targets to treat hepatic inflammation in general, and during parasite infection in particular.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Signal Transduction/immunology , Trypanosomiasis/immunology , Animals , Cell Separation , Chemokine CCL2/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Gene Expression Profiling , Interferon-gamma/immunology , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/immunology , Myeloid Differentiation Factor 88/immunology , Receptors, CCR2/immunology , Reverse Transcriptase Polymerase Chain Reaction , Trypanosoma brucei brucei/immunology
SELECTION OF CITATIONS
SEARCH DETAIL