Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 229(3): 888-897, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-37721470

ABSTRACT

BACKGROUND: Current tuberculosis treatment regimens could be improved by adjunct host-directed therapies (HDT) targeting host responses. We investigated the antimycobacterial capacity of macrophages from patients with tuberculosis in a phase 1/2 randomized clinical trial (TBCOX2) of the cyclooxygenase-2 inhibitor etoricoxib. METHODS: Peripheral blood mononuclear cells from 15 patients with tuberculosis treated with adjunctive COX-2i and 18 controls (standard therapy) were collected on day 56 after treatment initiation. The ex vivo capacity of macrophages to control mycobacterial infection was assessed by challenge with Mycobacterium avium, using an in vitro culture model. Macrophage inflammatory responses were analyzed by gene expression signatures, and concentrations of cytokines were analyzed in supernatants by multiplex. RESULTS: Macrophages from patients receiving adjunctive COX-2i treatment had higher M. avium loads than controls after 6 days, suggesting an impaired capacity to control mycobacterial infection compared to macrophages from the control group. Macrophages from the COX-2i group had lower gene expression of TNF, IL-1B, CCL4, CXCL9, and CXCL10 and lowered production of cytokines IFN-ß and S100A8/A9 than controls. CONCLUSIONS: Our data suggest potential unfavorable effects with impaired macrophage capacity to control mycobacterial growth in patients with tuberculosis receiving COX-2i treatment. Larger clinical trials are required to analyze the safety of COX-2i as HDT in patients with tuberculosis. CLINICAL TRIALS REGISTRATION: NCT02503839.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Cytokines , Etoricoxib/pharmacology , Leukocytes, Mononuclear , Macrophages/microbiology , Tuberculosis/microbiology
2.
Nanomedicine ; 54: 102712, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37838100

ABSTRACT

Nanomedicine holds immense potential for therapeutic manipulation of phagocytic immune cells. However, in vitro studies often fail to accurately translate to the complex in vivo environment. To address this gap, we employed an ex vivo human whole-blood assay to evaluate liposome interactions with immune cells. We systematically varied liposome size, PEG-surface densities and sphingomyelin and ganglioside content. We observed differential uptake patterns of the assessed liposomes by neutrophils and monocytes, emphasizing the importance of liposome design. Interestingly, our results aligned closely with published in vivo observations in mice and patients. Moreover, liposome exposure induced changes in cytokine release and cellular responses, highlighting the potential modulation of immune system. Our study highlights the utility of human whole-blood models in assessing nanoparticle-immune cell interactions and provides insights into liposome design for modulating immune responses.


Subject(s)
Liposomes , Phagocytes , Humans , Animals , Mice , Monocytes , Sphingomyelins , Cytokines
3.
Front Immunol ; 13: 891475, 2022.
Article in English | MEDLINE | ID: mdl-35874747

ABSTRACT

Macrophages deploy a variety of antimicrobial programs to contain mycobacterial infection. Upon activation, they undergo extensive metabolic reprogramming to meet an increase in energy demand, but also to support immune effector functions such as secretion of cytokines and antimicrobial activities. Here, we report that mitochondrial import of pyruvate is linked to production of mitochondrial ROS and control of Mycobacterium avium (M. avium) infection in human primary macrophages. Using chemical inhibition, targeted mass spectrometry and single cell image analysis, we showed that macrophages infected with M. avium switch to aerobic glycolysis without any major imbalances in the tricarboxylic acid cycle volume or changes in the energy charge. Instead, we found that pyruvate import contributes to hyperpolarization of mitochondria in infected cells and increases production of mitochondrial reactive oxygen species by the complex I via reverse electron transport, which reduces the macrophage burden of M. avium. While mycobacterial infections are extremely difficult to treat and notoriously resistant to antibiotics, this work stresses out that compounds specifically inducing mitochondrial reactive oxygen species could present themself as valuable adjunct treatments.


Subject(s)
Mycobacterium Infections , Mycobacterium avium-intracellulare Infection , Humans , Macrophages , Mitochondria/metabolism , Mycobacterium Infections/metabolism , Mycobacterium avium/physiology , Proto-Oncogene Proteins c-ret/metabolism , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
4.
J Alzheimers Dis ; 84(4): 1781-1794, 2021.
Article in English | MEDLINE | ID: mdl-34719495

ABSTRACT

BACKGROUND: Preclinical models of Alzheimer's disease (AD) can provide valuable insights into the onset and progression of the disease, such as changes in concentrations of amyloid-ß (Aß) and tau in cerebrospinal fluid (CSF). However, such models are currently underutilized due to limited advancement in techniques that allow for longitudinal CSF monitoring. OBJECTIVE: An elegant way to understand the biochemical environment in the diseased brain is intracerebral microdialysis, a method that has until now been limited to short-term observations, or snapshots, of the brain microenvironment. Here we draw upon patient-based findings to characterize CSF biomarkers in a commonly used preclinical mouse model for AD. METHODS: Our modified push-pull microdialysis method was first validated ex vivo with human CSF samples, and then in vivo in an AD mouse model, permitting assessment of dynamic changes of CSF Aß and tau and allowing for better translational understanding of CSF biomarkers. RESULTS: We demonstrate that CSF biomarker changes in preclinical models capture what is observed in the brain; with a decrease in CSF Aß observed when plaques are deposited, and an increase in CSF tau once tau pathology is present in the brain parenchyma. We found that a high molecular weight cut-off membrane allowed for simultaneous sampling of Aß and tau, comparable to CSF collection by lumbar puncture in patients. CONCLUSION: Our approach can further advance AD and other neurodegenerative research by following evolving neuropathology along the disease cascade via consecutive sampling from the same animal and can additionally be used to administer pharmaceutical compounds and assess their efficacy.


Subject(s)
Alzheimer Disease , Biomarkers/cerebrospinal fluid , Microdialysis , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Brain/pathology , Disease Models, Animal , Humans , Mice , tau Proteins/cerebrospinal fluid
5.
mBio ; 12(5): e0212121, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34607464

ABSTRACT

Macrophages sense and respond to pathogens by induction of antimicrobial and inflammatory programs to alert other immune cells and eliminate the infectious threat. We have previously identified the transcription factor IRF1 to be consistently activated in macrophages during Mycobacterium avium infection, but its precise role during infection is not clear. Here, we show that tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) autocrine/paracrine signaling contributes to controlling the intracellular growth of M. avium in human primary macrophages through activation of IRF1 nuclear translocation and expression of IRG1, a mitochondrial enzyme that produces the antimicrobial metabolite itaconate. Small interfering RNA (siRNA)-mediated knockdown of IRF1 or IRG1 increased the mycobacterial load, whereas exogenously provided itaconate was bacteriostatic at high concentrations. While the overall level of endogenous itaconate was low in M. avium-infected macrophages, the repositioning of mitochondria to M. avium phagosomes suggests a mechanism by which itaconate can be delivered directly to M. avium phagosomes in sufficient quantities to inhibit growth. Using mRNA hybridization, we further show that uninfected bystander cells actively contribute to the resolution of infection by producing IL-6 and TNF-α, which, via paracrine signaling, activate IRF1/IRG1 and strengthen the antimicrobial activity of infected macrophages. This mechanism contributes to the understanding of why patients on anti-inflammatory treatment, e.g., with tocilizumab or infliximab, can be more susceptible to mycobacterial disease. IMPORTANCE The prevalence of lung diseases caused by nontuberculous mycobacteria, such as Mycobacterium avium, is increasing in countries where tuberculosis is not endemic, most likely because of an aging population that is immunocompromised from underlying disease or immunosuppressive therapy. Our study contributes to the understanding of mycobacterial survival and killing in human macrophages and, more broadly, to the impact of immunometabolism during infection. We show evidence of an antimicrobial program in human primary macrophages where activation of the transcription factor IRF1 and expression of the mitochondrial enzyme IRG1 restrict the intracellular growth of M. avium, possibly by directed delivery of itaconate to M. avium phagosomes. The study also sheds light on why patients on immunosuppressive therapy are more susceptible to mycobacterial infections, since TNF-α and IL-6 contribute to driving the described antimycobacterial program.


Subject(s)
Carboxy-Lyases/immunology , Interferon Regulatory Factor-1/immunology , Interleukin-6/immunology , Macrophages/immunology , Mycobacterium avium/physiology , Tuberculosis/immunology , Tuberculosis/microbiology , Tumor Necrosis Factor-alpha/immunology , Carboxy-Lyases/genetics , Cells, Cultured , Humans , Immunity, Innate , Interferon Regulatory Factor-1/genetics , Macrophages/microbiology , Mycobacterium avium/genetics , Paracrine Communication , Tuberculosis/genetics , Tumor Necrosis Factor-alpha/genetics
6.
J Leukoc Biol ; 109(1): 23-33, 2021 01.
Article in English | MEDLINE | ID: mdl-32531827

ABSTRACT

Mycobacterium avium (Mav) causes chronic infections in immunocompromised patients that require long-term antibiotic treatment. We have previously shown that Mav takes residence in host Mϕs and establishes a compartment (MavC) in which it is hidden from host defenses. Failure to establish the MavC traps Mav in Lamp1+ phagolysosomes where growth is prevented, and inflammatory signaling activated through TLRs 7/8. To elucidate how antibiotic treatment affects mycobacterial trafficking and host defenses, we infected human primary Mϕs with Mav for 4 days prior to treatment with a macrolide, aminoglycoside, and ethambutol. We show that Mav is killed and the MavC fuses with Lamp1+ lysosomes following antibiotic treatment. However, this does not result in nuclear translocation of NF-κB or production of inflammatory cytokines, suggesting different Lamp1+ lysosomal compartments can form that differ in their innate signaling capabilities. Thus, we show that upon antibiotic treatment of a chronic infection, Mav is quietly disposed of by Mϕs.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Host-Pathogen Interactions/drug effects , Macrophages/drug effects , Macrophages/microbiology , Mycobacterium avium-intracellulare Infection , Cytokines/biosynthesis , Host-Pathogen Interactions/immunology , Humans , Macrophages/immunology , Mycobacterium avium Complex/drug effects , Phagosomes/metabolism , Phagosomes/microbiology
7.
Nat Commun ; 11(1): 147, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31919342

ABSTRACT

During HIV infection, cell-to-cell transmission results in endosomal uptake of the virus by target CD4+ T cells and potential exposure of the viral ssRNA genome to endosomal Toll-like receptors (TLRs). TLRs are instrumental in activating inflammatory responses in innate immune cells, but their function in adaptive immune cells is less well understood. Here we show that synthetic ligands of TLR8 boosted T cell receptor signaling, resulting in increased cytokine production and upregulation of surface activation markers. Adjuvant TLR8 stimulation, but not TLR7 or TLR9, further promoted T helper cell differentiation towards Th1 and Th17. In addition, we found that endosomal HIV induced cytokine secretion from CD4+ T cells in a TLR8-specific manner. TLR8 engagement also enhanced HIV-1 replication and potentiated the reversal of latency in patient-derived T cells. The adjuvant TLR8 activity in T cells can contribute to viral dissemination in the lymph node and low-grade inflammation in HIV patients. In addition, it can potentially be exploited for therapeutic targeting and vaccine development.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Toll-Like Receptor 8/metabolism , Cell Line , HIV Infections/transmission , Humans , Immunity, Innate/immunology , Signal Transduction/immunology , Toll-Like Receptor 8/immunology
8.
Biol Open ; 7(8)2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30026265

ABSTRACT

During early mammalian development, transient pools of pluripotent cells emerge that can be immortalised upon stem cell derivation. The pluripotent state, 'naïve' or 'primed', depends on the embryonic stage and derivation conditions used. Here we analyse the temporal gene expression patterns of mouse, cattle and porcine embryos at stages that harbour different types of pluripotent cells. We document conserved and divergent traits in gene expression, and identify predictor genes shared across the species that are associated with pluripotent states in vivo and in vitro Amongst these are the pluripotency-linked genes Klf4 and Lin28b The novel genes discovered include naïve- (Spic, Scpep1 and Gjb5) and primed-associated (Sema6a and Jakmip2) genes as well as naïve to primed transition genes (Dusp6 and Trip6). Both Gjb5 and Dusp6 play a role in pluripotency since their knockdown results in differentiation and downregulation of key pluripotency genes. Our interspecies comparison revealed new insights of pluripotency, pluripotent stem cell identity and a new molecular criterion for distinguishing between pluripotent states in various species, including human.

9.
PLoS Pathog ; 13(11): e1006712, 2017 11.
Article in English | MEDLINE | ID: mdl-29112990

ABSTRACT

[This corrects the article DOI: 10.1371/journal.ppat.1006551.].

10.
PLoS Pathog ; 13(8): e1006551, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28806745

ABSTRACT

Pathogenic mycobacteria reside in macrophages where they avoid lysosomal targeting and degradation through poorly understood mechanisms proposed to involve arrest of phagosomal maturation at an early endosomal stage. A clear understanding of how this relates to host defenses elicited from various intracellular compartments is also missing and can only be studied using techniques allowing single cell and subcellular analyses. Using confocal imaging of human primary macrophages infected with Mycobacterium avium (Mav) we show evidence that Mav phagosomes are not arrested at an early endosomal stage, but mature to a (LAMP1+/LAMP2+/CD63+) late endosomal/phagolysosomal stage where inflammatory signaling and Mav growth restriction is initiated through a mechanism involving Toll-like receptors (TLR) 7 and 8, the adaptor MyD88 and transcription factors NF-κB and IRF-1. Furthermore, a fraction of the mycobacteria re-establish in a less hostile compartment (LAMP1-/LAMP2-/CD63-) where they not only evade destruction, but also recognition by TLRs, growth restriction and inflammatory host responses that could be detrimental for intracellular survival and establishment of chronic infections.


Subject(s)
Macrophages/microbiology , Mycobacterium Infections/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 7/immunology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Lysosomes/immunology , Macrophages/immunology , Microscopy, Confocal , Mycobacterium avium , Phagosomes/immunology , Polymerase Chain Reaction
11.
Proc Natl Acad Sci U S A ; 112(31): E4272-80, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195781

ABSTRACT

Several mechanisms are involved in controlling intracellular survival of pathogenic mycobacteria in host macrophages, but how these mechanisms are regulated remains poorly understood. We report a role for Kelch-like ECH-associated protein 1 (Keap1), an oxidative stress sensor, in regulating inflammation induced by infection with Mycobacterium avium in human primary macrophages. By using confocal microscopy, we found that Keap1 associated with mycobacterial phagosomes in a time-dependent manner, whereas siRNA-mediated knockdown of Keap1 increased M. avium-induced expression of inflammatory cytokines and type I interferons (IFNs). We show evidence of a mechanism whereby Keap1, as part of an E3 ubiquitin ligase complex with Cul3 and Rbx1, facilitates ubiquitination and degradation of IκB kinase (IKK)-ß thus terminating IKK activity. Keap1 knockdown led to increased nuclear translocation of transcription factors NF-κB, IFN regulatory factor (IRF) 1, and IRF5 driving the expression of inflammatory cytokines and IFN-ß. Furthermore, knockdown of other members of the Cul3 ubiquitin ligase complex also led to increased cytokine expression, further implicating this ligase complex in the regulation of the IKK family. Finally, increased inflammatory responses in Keap1-silenced cells contributed to decreased intracellular growth of M. avium in primary human macrophages that was reconstituted with inhibitors of IKKß or TANK-binding kinase 1 (TBK1). Taken together, we propose that Keap1 acts as a negative regulator for the control of inflammatory signaling in M. avium-infected human primary macrophages. Although this might be important to avoid sustained or overwhelming inflammation, our data suggest that a negative consequence could be facilitated growth of pathogens like M. avium inside macrophages.


Subject(s)
Inflammation/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium avium/physiology , Signal Transduction , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cytokines/biosynthesis , Gene Knockdown Techniques , Humans , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/metabolism , Kelch-Like ECH-Associated Protein 1 , Mycobacterium avium/growth & development , NF-kappa B/metabolism , Phagosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Protein Transport , Proteolysis , Reactive Oxygen Species/metabolism , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/pathology , Ubiquitination , Up-Regulation
12.
Nat Chem Biol ; 10(9): 707-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25064832

ABSTRACT

The PTH receptor is to our knowledge one of the first G protein-coupled receptor (GPCR) found to sustain cAMP signaling after internalization of the ligand-receptor complex in endosomes. This unexpected model is adding a new dimension on how we think about GPCR signaling, but its mechanism is incompletely understood. We report here that endosomal acidification mediated by the PKA action on the v-ATPase provides a negative feedback mechanism by which endosomal receptor signaling is turned off.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Endosomes/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Vacuolar Proton-Translocating ATPases/physiology , Arrestins/chemistry , Arrestins/metabolism , Cholera Toxin/pharmacology , Cyclic AMP/physiology , Feedback, Physiological , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Phosphorylation , Protein Binding , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 1/physiology , beta-Arrestins
13.
Mol Reprod Dev ; 79(7): 461-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22573702

ABSTRACT

Pluripotency can be captured in vitro, providing that the culture environment meets the requirements that avoid differentiation while stimulating self-renewal. From studies in the mouse embryo, two kinds of pluripotent stem cells have been obtained from the early and late epiblast, embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs), representing the naive and primed states, respectively. All attempts to derive convincing ESCs in ungulates have been unsuccessful, although all attempts were based on the assumption that the conditions used to derive mouse ESCs or human ESC could be applied in other species. Pluripotent cells derived in primates, rabbit, and pig strongly indicate that the state of pluripotency of these cells is, in fact, closer to EpiSCs than to ESCs, and thus depend on fibroblast growth factor (FGF) and Activin signaling pathways. Based on this observation, we have tried to derive EpiSC from the epiblast of bovine elongated embryos as well as ESCs from Day-8 blastocysts. We here show that the core transcription factors Oct4/Sox2/Nanog can be used as markers of pluripotency in the bovine since their expression was restricted to the developing epiblast after Day 8, and disappeared following differentiation of both the ESC-like and EpiSC-like cultures. Although FGF and Activin pathways are indeed present and active in the bovine, it is not sufficient/enough to maintain a long-term pluripotency ex vivo, as was reported for mouse and pig EpiSCs.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Germ Layers/metabolism , Pluripotent Stem Cells/metabolism , Activins/metabolism , Animals , Biomarkers , Blastocyst/metabolism , Cattle , Cells, Cultured , Embryonic Stem Cells/cytology , Fibroblast Growth Factors/metabolism , Germ Layers/cytology , Homeodomain Proteins/biosynthesis , Mice , Octamer Transcription Factor-3/biosynthesis , Pluripotent Stem Cells/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/biosynthesis , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...