Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Immunity ; 33(4): 583-96, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20933442

ABSTRACT

Toll-like receptor 4 (TLR4) is indispensable for recognition of Gram-negative bacteria. We described a trafficking pathway for TLR4 from the endocytic recycling compartment (ERC) to E. coli phagosomes. We found a prominent colocalization between TLR4 and the small GTPase Rab11a in the ERC, and Rab11a was involved in the recruitment of TLR4 to phagosomes in a process requiring TLR4 signaling. Also, Toll-receptor-associated molecule (TRAM) and interferon regulatory factor-3 (IRF3) localized to E. coli phagosomes and internalization of E. coli was required for a robust interferon-ß induction. Suppression of Rab11a reduced TLR4 in the ERC and on phagosomes leading to inhibition of the IRF3 signaling pathway induced by E. coli, whereas activation of the transcription factor NF-κB was unaffected. Moreover, Rab11a silencing reduced the amount of TRAM on phagosomes. Thus, Rab11a is an important regulator of TLR4 and TRAM transport to E. coli phagosomes thereby controlling IRF3 activation from this compartment.


Subject(s)
Phagosomes/metabolism , Toll-Like Receptor 4/physiology , rab GTP-Binding Proteins/physiology , Endocytosis , Escherichia coli/immunology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Phagocytosis , Signal Transduction , Staphylococcus aureus/immunology
2.
Biomed Microdevices ; 20(1): 9, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29294210

ABSTRACT

Neural network formation is a complex process involving axon outgrowth and guidance. Axon guidance is facilitated by structural and molecular cues from the surrounding microenvironment. Micro-fabrication techniques can be employed to produce microfluidic chips with a highly controlled microenvironment for neural cells enabling longitudinal studies of complex processes associated with network formation. In this work, we demonstrate a novel open microfluidic chip design that encompasses a freely variable number of nodes interconnected by axon-permissible tunnels, enabling structuring of multi-nodal neural networks in vitro. The chip employs a partially open design to allow high level of control and reproducibility of cell seeding, while reducing shear stress on the cells. We show that by culturing dorsal root ganglion cells (DRGs) in our microfluidic chip, we were able to structure a neural network in vitro. These neurons were compartmentalized within six nodes interconnected through axon growth tunnels. Furthermore, we demonstrate the additional benefit of open top design by establishing a 3D neural culture in matrigel and a neuronal aggregate 3D culture within the chips. In conclusion, our results demonstrate a novel microfluidic chip design applicable to structuring complex neural networks in vitro, thus providing a versatile, highly relevant platform for the study of neural network dynamics applicable to developmental and regenerative neuroscience.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Lab-On-A-Chip Devices , Nerve Net/cytology , Animals , Collagen , Drug Combinations , Equipment Design , Ganglia, Spinal/cytology , Laminin , Neurons/cytology , Proteoglycans , Rats, Sprague-Dawley
3.
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
4.
Immunology ; 140(2): 232-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23746054

ABSTRACT

Opportunistic infections with non-tuberculous mycobacteria such as Mycobacterium avium are receiving renewed attention because of increased incidence and difficulties in treatment. As for other mycobacterial infections, a still poorly understood collaboration of different immune effector mechanisms is required to confer protective immunity. Here we have characterized the interplay of innate and adaptive immune effector mechanisms contributing to containment in a mouse infection model using virulent M. avium strain 104 in C57BL/6 mice. M. avium caused chronic infection in mice, as shown by sustained organ bacterial load. In the liver, bacteria were contained in granuloma-like structures that could be defined morphologically by expression of the antibacterial innate effector protein Lipocalin 2 in the adjoining hepatocytes and infiltrating neutrophils, possibly contributing to containment. Circulatory anti-mycobacterial antibodies steadily increased throughout infection and were primarily of the IgM isotype. Highest levels of interferon-γ were found in infected liver, spleen and serum of mice approximately 2 weeks post infection and coincided with a halt in organ bacterial growth. In contrast, expression of tumour necrosis factor was surprisingly low in spleen compared with liver. We did not detect interleukin-17 in infected organs or M. avium-specific T helper 17 cells, suggesting a minor role for T helper 17 cells in this model. A transient and relative decrease in regulatory T cell numbers was seen in spleens. This detailed characterization of M. avium infection in C57BL/6 mice may provide a basis for future studies aimed at gaining better insight into mechanisms leading to containment of infections with non-tuberculous mycobacteria.


Subject(s)
Tuberculosis/immunology , Tuberculosis/pathology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mycobacterium avium , Real-Time Polymerase Chain Reaction
5.
Mater Today Bio ; 18: 100537, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36659998

ABSTRACT

Cell migration is essential in numerous living processes, including embryonic development, wound healing, immune responses, and cancer metastasis. From individual cells to collectively migrating epithelial sheets, the locomotion of cells is tightly regulated by multiple structural, chemical, and biological factors. However, the high complexity of this process limits the understanding of the influence of each factor. Recent advances in materials science, tissue engineering, and microtechnology have expanded the toolbox and allowed the development of biomimetic in vitro assays to investigate the mechanisms of cell migration. Particularly, three-dimensional (3D) hydrogels have demonstrated a superior ability to mimic the extracellular environment. They are therefore well suited to studying cell migration in a physiologically relevant and more straightforward manner than in vivo approaches. A myriad of synthetic and naturally derived hydrogels with heterogeneous characteristics and functional properties have been reported. The extensive portfolio of available hydrogels with different mechanical and biological properties can trigger distinct biological responses in cells affecting their locomotion dynamics in 3D. Herein, we describe the most relevant hydrogels and their associated physico-chemical characteristics typically employed to study cell migration, including established cell migration assays and tracking methods. We aim to give the reader insight into existing literature and practical details necessary for performing cell migration studies in 3D environments.

6.
J Immunol ; 182(1): 588-95, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109192

ABSTRACT

Soluble proteins that bind LPS, like myeloid differentiation-2 (MD-2) and CD14, have essential roles in regulating LPS signaling through TLR4. During a gram-negative bacterial infection, the host may control the response by adjusting the levels of soluble MD-2 and CD14. To address the surface expression of MD-2 on human leukocytes, we developed a mAb, IIC1, that recognized MD-2 both free and when bound to TLR4. MD-2 was found on the surface of freshly isolated monocytes, on a subpopulation of CD19(+) B-cells and on CD15(+) neutrophils. LPS transiently reduced the MD-2 levels on monocytes, which is most likely due to endocytosis of the LPS receptor complex since MD-2 colocalized with TLR4 in early endosomes after LPS stimulation. In the absence of LPS, MD-2 partly colocalized with TLR4 in Golgi trans and medial compartments. Cultivating monocytes for 18-20 h resulted in loss of MD-2 expression on the surface, which was reversed either by LPS or IL-10. Furthermore, addition of IL-10, but not LPS, resulted in a considerable increase in mRNA for both MD-2 and CD14. Using ELISA, we demonstrated that IL-10 had a profound dose- and time-related effect on the release of soluble MD-2 and soluble CD14 from monocytes. In HIV-infected patients, the amounts of MD-2, CD14, and IL-10 increased significantly in the patient group with AIDS. Of interest, we found that IL-10, CD14, and MD-2 levels were positively correlated, suggesting that IL-10 may be a driving force for increased release of MD-2 and CD14 during systemic inflammation.


Subject(s)
HIV Infections/immunology , HIV Infections/metabolism , Interleukin-10/physiology , Lipopolysaccharide Receptors/biosynthesis , Lymphocyte Antigen 96/biosynthesis , Monocytes/immunology , Monocytes/metabolism , Up-Regulation/immunology , Adult , Animals , Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , CHO Cells , Cell Line , Cricetinae , Cricetulus , Female , Humans , Inflammation Mediators/physiology , Interleukin-10/blood , Lipopolysaccharide Receptors/blood , Lipopolysaccharide Receptors/genetics , Lymphocyte Antigen 96/blood , Lymphocyte Antigen 96/genetics , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/biosynthesis
7.
J Infect Dis ; 201(5): 783-92, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20121435

ABSTRACT

Iron is an essential nutrient for microbes, and many pathogenic bacteria depend on siderophores to obtain iron. The mammalian innate immunity protein lipocalin 2 (Lcn2; also known as neutrophil gelatinase-associated lipocalin, 24p3, or siderocalin) binds the siderophore carboxymycobactin, an essential component of the iron acquisition apparatus of mycobacteria. Here we show that Lcn2 suppressed growth of Mycobacterium avium in culture, and M. avium induced Lcn2 production from mouse macrophages. Lcn2 also had elevated levels and initially limited the growth of M. avium in the blood of infected mice but did not impede growth in tissues and during long-term infections. M. avium is an intracellular pathogen. Subcellular imaging of infected macrophages revealed that Lcn2 trafficked to lysosomes separate from M. avium, whereas transferrin was efficiently transported to the mycobacteria. Thus, mycobacteria seem to reside in the Rab11(+) endocytic recycling pathway, thereby retaining access to nutrition and avoiding endocytosed immunoproteins like Lcn2.


Subject(s)
Acute-Phase Proteins/immunology , Acute-Phase Proteins/metabolism , Lipocalins/immunology , Lipocalins/metabolism , Lysosomes/metabolism , Lysosomes/microbiology , Mycobacterium avium/immunology , Mycobacterium avium/pathogenicity , Oncogene Proteins/immunology , Oncogene Proteins/metabolism , Transferrin/metabolism , Animals , Blood/microbiology , Colony Count, Microbial , Lipocalin-2 , Lipocalins/blood , Liver/microbiology , Lysosomes/chemistry , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium avium/growth & development , Mycobacterium avium/metabolism , Oncogene Proteins/blood , Spleen/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology , rab GTP-Binding Proteins/metabolism
8.
Biosens Bioelectron ; 140: 111329, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31163396

ABSTRACT

Lab-on-chip platforms, such as microfluidic chips and micro-electrode arrays (MEAs) are powerful tools that allow us to manipulate and study neurons in vitro. Microfluidic chips provide a controlled extracellular environment that structures neural networks and facilitates isolation and manipulation at a sub-cellular level. Furthermore, MEAs enable measurement of extracellular electrophysiological activity from single neurons to entire networks. Here, we demonstrate the design, fabrication and application of a 3-nodal microfluidic chip integrated with MEAs as a versatile study platform for neurobiology and pathophysiology. In this work, we evaluate the use of the microfluidic chip to structure a neural network into three separate nodes, interconnected through tunnels that isolate and guide axons into a channel, thus facilitating synaptic contacts between neurons originating from opposite nodes. Furthermore, we demonstrate the utility of the MEA for monitoring developing activity and intra-/inter nodal connectivity of the structured neural network. Finally, we demonstrate the versatility of the platform in two separate experiments. First, we demonstrate the ability to measure intra- and inter-nodal dynamic responses to a fluidically isolated chemical stimulation. Then, we demonstrate the feature of the microfluidic chip enabling the disruption of functional connectivity between nodes and examination of the immediate activity response of the neural network. The platform enables in vitro modelling of neural networks to study their functional connectomes in the context of neurodegenerative disease and CNS trauma, including spinal cord injury.


Subject(s)
Biosensing Techniques/instrumentation , Lab-On-A-Chip Devices , Nerve Net/cytology , Nerve Net/drug effects , Neurotransmitter Agents/pharmacology , Animals , Axotomy , Cell Line , Equipment Design , Nerve Net/physiology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Synapses/drug effects , Synapses/metabolism
9.
Front Immunol ; 9: 650, 2018.
Article in English | MEDLINE | ID: mdl-29670624

ABSTRACT

Effective priming and activation of tumor-specific CD8+ cytotoxic T lymphocytes (CTLs) is crucial for realizing the potential of therapeutic cancer vaccination. This requires cytosolic antigens that feed into the MHC class I presentation pathway, which is not efficiently achieved with most current vaccination technologies. Photochemical internalization (PCI) provides an emerging technology to route endocytosed material to the cytosol of cells, based on light-induced disruption of endosomal membranes using a photosensitizing compound. Here, we investigated the potential of PCI as a novel, minimally invasive, and well-tolerated vaccination technology to induce priming of cancer-specific CTL responses to peptide antigens. We show that PCI effectively promotes delivery of peptide antigens to the cytosol of antigen-presenting cells (APCs) in vitro. This resulted in a 30-fold increase in MHC class I/peptide complex formation and surface presentation, and a subsequent 30- to 100-fold more efficient activation of antigen-specific CTLs compared to using the peptide alone. The effect was found to be highly dependent on the dose of the PCI treatment, where optimal doses promoted maturation of immature dendritic cells, thus also providing an adjuvant effect. The effect of PCI was confirmed in vivo by the successful induction of antigen-specific CTL responses to cancer antigens in C57BL/6 mice following intradermal peptide vaccination using PCI technology. We thus show new and strong evidence that PCI technology holds great potential as a novel strategy for improving the outcome of peptide vaccines aimed at triggering cancer-specific CD8+ CTL responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunotherapy/methods , Neoplasms/therapy , Vaccination/methods , Animals , Antigen Presentation , Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic , Endocytosis , Humans , Injections, Intradermal , Mice , Neoplasms/immunology , Peptides/immunology , Photochemical Processes
10.
Lab Chip ; 16(19): 3728-40, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27560793

ABSTRACT

T cells play a central role in immunity towards cancer and infectious diseases. T cell responses are initiated in the T cell zone of the lymph node (LN), where resident antigen-bearing dendritic cells (DCs) prime and activate antigen-specific T cells passing by. In the present study, we investigated the T cell : DC interaction in a microfluidic device to understand the intercellular dynamics and physiological conditions in the LN. We show random migration of antigen-specific T cells onto the antigen-presenting DC monolayer independent of the flow direction with a mean T cell : DC dwell time of 12.8 min and a mean velocity of 6 µm min(-1). Furthermore, we investigated the antigen specific vs. unspecific attachment and detachment of CD8(+) and CD4(+) T cells to DCs under varying shear stress. In our system, CD4(+) T cells showed long stable contacts with APCs, whereas CD8(+) T cells presented transient interactions with DCs. By varying the shear stress from 0.01 to 100 Dyn cm(-2), it was also evident that there was a much stronger attachment of antigen-specific than unspecific T cells to stationary DCs up to 1-12 Dyn cm(-2). The mechanical force of the cell : cell interaction associated with the pMHC-TCR match under controlled tangential shear force was estimated to be in the range of 0.25-4.8 nN. Finally, upon performing attachment & detachment tests, there was a steady accumulation of antigen specific CD8(+) T cells and CD4(+) T cells on DCs at low shear stresses, which were released at a stress of 12 Dyn cm(-2). This microphysiological model provides new possibilities to recreate a controlled mechanical force threshold of pMHC-TCR binding, allowing the investigation of intercellular signalling of immune synapses and therapeutic targets for immunotherapy.


Subject(s)
Cell Communication , Dendritic Cells/cytology , Lab-On-A-Chip Devices , Lymph Nodes/immunology , T-Lymphocytes/cytology , Animals , Lymphocyte Activation , Mice , Shear Strength , Stress, Mechanical , T-Lymphocytes/immunology
11.
PLoS One ; 10(9): e0134644, 2015.
Article in English | MEDLINE | ID: mdl-26406896

ABSTRACT

Mycobacteria pose a threat to the world health today, with pathogenic and opportunistic bacteria causing tuberculosis and non-tuberculous disease in large parts of the population. Much is still unknown about the interplay between bacteria and host during infection and disease, and more research is needed to meet the challenge of drug resistance and inefficient vaccines. This work establishes a reliable and reproducible method for performing correlative imaging of human macrophages infected with mycobacteria at an ultra-high resolution and in 3D. Focused Ion Beam/Scanning Electron Microscopy (FIB/SEM) tomography is applied, together with confocal fluorescence microscopy for localization of appropriately infected cells. The method is based on an Aclar poly(chloro-tri-fluoro)ethylene substrate, micropatterned into an advantageous geometry by a simple thermomoulding process. The platform increases the throughput and quality of FIB/SEM tomography analyses, and was successfully applied to detail the intracellular environment of a whole mycobacterium-infected macrophage in 3D.


Subject(s)
Imaging, Three-Dimensional , Leukocytes, Mononuclear/microbiology , Leukocytes, Mononuclear/pathology , Mycobacterium , Electron Microscope Tomography , Humans , Image Processing, Computer-Assisted , Leukocytes, Mononuclear/ultrastructure , Macrophages/microbiology , Macrophages/pathology , Macrophages/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Scanning , Mycobacterium/physiology
13.
EMBO J ; 25(4): 683-92, 2006 Feb 22.
Article in English | MEDLINE | ID: mdl-16467847

ABSTRACT

Immune responses are initiated when molecules of microbial origin are sensed by the Toll-like receptors (TLRs). We now report the identification of essential molecular components for the trafficking of the lipopolysaccharide (LPS) receptor complex. LPS was endocytosed by a receptor-mediated mechanism dependent on dynamin and clathrin and colocalized with TLR4 on early/sorting endosomes. TLR4 was ubiquitinated and associated with the ubiquitin-binding endosomal sorting protein hepatocyte growth factor-regulated tyrosine kinase substrate, Hrs. Inhibition of endocytosis and endosomal sorting increased LPS signaling. Finally, the LPS receptor complex was sorted to late endosomes/lysosomes for degradation and loading of associated antigens onto HLA class II molecules for presentation to CD4+ T cells. Our results show that endosomal trafficking of the LPS receptor complex is essential for signal termination and LPS-associated antigen presentation, thus controlling both innate and adaptive immunity through TLR4.


Subject(s)
Antigen Presentation/immunology , Endocytosis/immunology , Immunity, Innate/immunology , Lipopolysaccharides/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Antigen Presentation/drug effects , Biological Transport, Active/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Clathrin/immunology , Dynamins/immunology , Endocytosis/drug effects , Endosomal Sorting Complexes Required for Transport , HLA Antigens/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Phosphoproteins/immunology , Signal Transduction/drug effects
14.
J Biol Chem ; 277(38): 35489-95, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12089142

ABSTRACT

The alginate capsule produced by the human pathogen Pseudomonas aeruginosa is composed mainly of mannuronic acid polymers (poly-M) that have immunostimulating properties. Poly-M shares with lipopolysaccharide the ability to stimulate cytokine production from human monocytes in a CD14-dependent manner. In the present study we examined the role of Toll-like receptor (TLR) 2 and TLR4 in responses to poly-M. Blocking antibodies to TLR2 and TLR4 partly inhibited tumor necrosis factor production induced by poly-M in human monocytes, and further inhibition was obtained by combining the antibodies. By transiently transfecting HEK293 cells, we found that membrane CD14 together with either TLR2 or TLR4/MD-2 could mediate activation by poly-M. Transfection of HEK293 cells with TLR2 and fluorescently labeled TLR4 followed by co-patching of TLR2 with an antibody revealed no association of these molecules on the plasma membrane. However, macrophages from the Tlr4 mutant C3H/HeJ mice and TLR4 knockout mice were completely non-responsive to poly-M, whereas the tumor necrosis factor release from TLR2 knockout macrophages was half of that seen with wild type cells. Taken together the results suggest that both TLR2 and TLR4 are involved in cell activation by poly-M and that TLR4 may be required in primary murine macrophages.


Subject(s)
Biopolymers/physiology , Drosophila Proteins , Hexuronic Acids/metabolism , Membrane Glycoproteins/physiology , Protein Isoforms/physiology , Receptors, Cell Surface/physiology , Animals , Cell Line , Cell Membrane/metabolism , Humans , Interleukin-6/biosynthesis , Mice , Mice, Inbred C3H , Signal Transduction , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL