Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.207
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 31: 743-91, 2013.
Article in English | MEDLINE | ID: mdl-23330953

ABSTRACT

Dendritic cells (DCs) are specialized sentinels responsible for coordinating adaptive immunity. This function is dependent upon coupled sensitivity to environmental signs of inflammation and infection to cellular maturation-the programmed alteration of DC phenotype and function to enhance immune cell activation. Although DCs are thus well equipped to respond to pathogens, maturation triggers are not unique to infection. Given that immune cells are exquisitely sensitive to the biological functions of DCs, we now appreciate that multiple layers of suppression are required to restrict the environmental sensitivity, cellular maturation, and even life span of DCs to prevent aberrant immune activation during the steady state. At the same time, steady-state DCs are not quiescent but rather perform key functions that support homeostasis of numerous cell types. Here we review these functions and molecular mechanisms of suppression that control steady-state DC maturation. Corruption of these steady-state operatives has diverse immunological consequences and pinpoints DCs as potent drivers of autoimmune and inflammatory disease.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Homeostasis/immunology , Signal Transduction/immunology , Animals , Cell Differentiation/genetics , Dendritic Cells/metabolism , Homeostasis/genetics , Humans , Lectins, C-Type/physiology , Membrane Glycoproteins/physiology , Mice , Receptors, Immunologic/physiology , Receptors, Pattern Recognition/physiology , Signal Transduction/genetics , Toll-Like Receptors/physiology
2.
Nat Immunol ; 15(7): 612-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24859449

ABSTRACT

Excessive activation of dendritic cells (DCs) leads to the development of autoimmune and inflammatory diseases, which has prompted a search for regulators of DC activation. Here we report that Rhbdd3, a member of the rhomboid family of proteases, suppressed the activation of DCs and production of interleukin 6 (IL-6) triggered by Toll-like receptors (TLRs). Rhbdd3-deficient mice spontaneously developed autoimmune diseases characterized by an increased abundance of the TH17 subset of helper T cells and decreased number of regulatory T cells due to the increase in IL-6 from DCs. Rhbdd3 directly bound to Lys27 (K27)-linked polyubiquitin chains on Lys302 of the modulator NEMO (IKKγ) via the ubiquitin-binding-association (UBA) domain in endosomes. Rhbdd3 further recruited the deubiquitinase A20 via K27-linked polyubiquitin chains on Lys268 to inhibit K63-linked polyubiquitination of NEMO and thus suppressed activation of the transcription factor NF-κB in DCs. Our data identify Rhbdd3 as a critical regulator of DC activation and indicate K27-linked polyubiquitination is a potent ubiquitin-linked pattern involved in the control of autoimmunity.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Autoimmunity , Dendritic Cells/immunology , Interleukin-6/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitination , Animals , Interleukin-6/antagonists & inhibitors , Lysine/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Protein Structure, Tertiary , T-Lymphocytes/immunology , Toll-Like Receptors/physiology
3.
Handb Exp Pharmacol ; 276: 291-314, 2022.
Article in English | MEDLINE | ID: mdl-34783909

ABSTRACT

Toll-like receptors (TLRs) receptors are responsible for initiation of inflammatory responses by their recognition of molecular patterns present in invading microorganisms (such as bacteria, viruses or fungi) or in molecules released following tissue damage in disease states. Expressed in the intestinal epithelium, they initiate an intracellular signalling cascade in response to molecular patterns resulting in the activation of transcription factors and the release of cytokines, chemokines and vasoactive molecules. Intestinal epithelial cells are exposed to microorganisms on a daily basis and form part of the primary defence against pathogens by using TLRs. TLRs and their accessory molecules are subject to tight regulation in these cells so as to not overreact or react in unnecessary circumstances. TLRs have more recently been associated with chronic inflammatory diseases as a result of inappropriate regulation, this can be damaging and lead to chronic inflammatory diseases such as inflammatory bowel disease (IBD). Targeting Toll-like receptors offers a potential therapeutic approach for IBD. In this review, the current knowledge on the TLRs is reviewed along with their association with intestinal diseases. Finally, compounds that target TLRs in animal models of IBD, clinic trials and their future merit as targets are discussed.


Subject(s)
Inflammatory Bowel Diseases , Toll-Like Receptors , Animals , Cytokines , Humans , Immunity, Innate , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa , Signal Transduction/physiology , Toll-Like Receptors/physiology , Toll-Like Receptors/therapeutic use
4.
IUBMB Life ; 73(7): 900-915, 2021 07.
Article in English | MEDLINE | ID: mdl-34033211

ABSTRACT

Toll-like receptors (TLRs) or pattern recognition receptors respond to pathogen-associated molecular patterns (PAMPs) or internal damage-associated molecular patterns (DAMPs). TLRs are integral membrane proteins with both extracellular leucine-rich and cytoplasmic domains that initiate downstream signaling through kinases by activating transcription factors like AP-1 and NF-κB, which lead to the release of various inflammatory cytokines and immune modulators. In the central nervous system, different TLRs are expressed mainly in microglia and astroglial cells, although some TLRs are also expressed in oligodendroglia and neurons. Activation of TLRs triggers signaling cascades by the host as a defense mechanism against invaders to repair damaged tissue. However, overactivation of TLRs disrupts the sustained immune homeostasis-induced production of pro-inflammatory molecules, such as cytokines, miRNAs, and inflammatory components of extracellular vesicles. These inflammatory mediators can, in turn, induce neuroinflammation, and neural tissue damage associated with many neurodegenerative diseases. This review discusses the critical role of TLRs response in Alzheimer's disease, Parkinson's disease, ischemic stroke, amyotrophic lateral sclerosis, and alcohol-induced brain damage and neurodegeneration.


Subject(s)
Alcoholism/physiopathology , Brain/drug effects , Neurodegenerative Diseases/etiology , Neuroinflammatory Diseases/etiology , Toll-Like Receptors/physiology , Alcoholism/etiology , Animals , Brain/physiopathology , Exosomes/pathology , Exosomes/physiology , Gene Expression , Humans , Immunity, Innate , MicroRNAs/genetics , MicroRNAs/metabolism , Neurodegenerative Diseases/therapy , Neuroinflammatory Diseases/therapy
5.
Nat Immunol ; 10(9): 965-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668221

ABSTRACT

The intensity and duration of immune responses are controlled by many proteins that modulate Toll-like receptor (TLR) signaling. TANK has been linked to positive regulation of the transcription factors IRF3 and NF-kappaB. Here we demonstrate that TANK is not involved in interferon responses and is a negative regulator of proinflammatory cytokine production induced by TLR signaling. TLR-induced polyubiquitination of the ubiquitin ligase TRAF6 was upregulated in Tank(-/-) macrophages. Notably, Tank(-/-) mice spontaneously developed fatal glomerulonephritis owing to deposition of immune complexes. Autoantibody production in Tank(-/-) mice was abrogated by antibiotic treatment or the absence of interleukin 6 (IL-6) or the adaptor MyD88. Our results demonstrate that constitutive TLR signaling by intestinal commensal microflora is suppressed by TANK.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Autoimmune Diseases/prevention & control , Glomerulonephritis/prevention & control , Signal Transduction/physiology , Toll-Like Receptors/physiology , Animals , Autoimmunity , CD40 Antigens/physiology , Female , Intestines/microbiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , Receptors, Antigen, B-Cell/physiology , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin/metabolism
6.
Nat Immunol ; 10(4): 340-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295630

ABSTRACT

An important function of receptors that signal through immunoreceptor tyrosine-based activation motifs (ITAMs) is to regulate signaling by heterologous receptors. This review describes mechanisms by which ITAM-associated receptors modulate signaling by Toll-like receptors (TLRs), tumor necrosis factor receptor family members and cytokine receptors that use the Jak-STAT signaling pathway, and the biological importance of this signal transduction cross-talk. ITAM-mediated cross-regulation can either augment or dampen signaling by other receptors. Conversely, TLRs and cytokines modulate ITAM-mediated signaling, by means including activation of beta2 integrins that are coupled to the ITAM-containing adaptors DAP12 and FcRgamma. Integration of ITAM signaling into signaling networks through cross-talk with other signal transduction pathways results in tight regulation and fine tuning of cellular responses to various extracellular stimuli and contributes to induction of specific activation and differentiation pathways.


Subject(s)
Amino Acid Motifs , Receptors, Cell Surface/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , CD18 Antigens/immunology , CD18 Antigens/metabolism , Humans , Immunoglobulin Variable Region/immunology , Janus Kinases/immunology , Janus Kinases/physiology , Membrane Proteins/immunology , Receptor Cross-Talk/immunology , Receptors, Cell Surface/immunology , Receptors, Cytokine/immunology , Receptors, Cytokine/physiology , Receptors, Immunologic/immunology , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , STAT Transcription Factors/immunology , STAT Transcription Factors/physiology , Toll-Like Receptors/immunology , Toll-Like Receptors/physiology
7.
Brain Behav Immun ; 91: 740-755, 2021 01.
Article in English | MEDLINE | ID: mdl-33039660

ABSTRACT

Central nervous system (CNS) innate immunity plays essential roles in infections, neurodegenerative diseases, and brain or spinal cord injuries. Astrocytes and microglia are the principal cells that mediate innate immunity in the CNS. Pattern recognition receptors (PRRs), expressed by astrocytes and microglia, sense pathogen-derived or endogenous ligands released by damaged cells and initiate the innate immune response. Toll-like receptors (TLRs) are a well-characterized family of PRRs. The contribution of microglial TLR signaling to CNS pathology has been extensively investigated. Even though astrocytes assume a wide variety of key functions, information about the role of astroglial TLRs in CNS disease and injuries is limited. Because astrocytes display heterogeneity and exhibit phenotypic plasticity depending on the effectors present in the local milieu, they can exert both detrimental and beneficial effects. TLRs are modulators of these paradoxical astroglial properties. The goal of the current review is to highlight the essential roles played by astroglial TLRs in CNS infections, injuries and diseases. We discuss the contribution of astroglial TLRs to host defense as well as the dissemination of viral and bacterial infections in the CNS. We examine the link between astroglial TLRs and the pathogenesis of neurodegenerative diseases and present evidence showing the pivotal influence of astroglial TLR signaling on sterile inflammation in CNS injury. Finally, we define the research questions and areas that warrant further investigations in the context of astrocytes, TLRs, and CNS dysfunction.


Subject(s)
Astrocytes/metabolism , Neurodegenerative Diseases/physiopathology , Toll-Like Receptors/physiology , Animals , Astrocytes/physiology , Brain/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System Diseases/immunology , Central Nervous System Infections/pathology , Encephalitis/immunology , Humans , Immunity, Innate/physiology , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Receptors, Pattern Recognition/immunology , Signal Transduction , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Toll-Like Receptors/metabolism
8.
Biosci Biotechnol Biochem ; 85(3): 656-665, 2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33589925

ABSTRACT

Macrophages can initiate innate immune responses against microbes and cancer. The aim of this study was to elucidate the effects of Brassica rapa L. on macrophages. The production of interleukin (IL)-6, tumor necrosis factor (TNF)-α, and interferon-γ induced by the insoluble fraction of B. rapa L. was decreased in macrophage-depleted spleen cells compared with controls. The insoluble fraction of B. rapa L. induced expression of H-2Kb, I-Ab, CD40, and CD86, production of cytokines and nitric oxide, and phagocytic activity in RAW264 cells. After treatment with the insoluble fraction, IL-6 and TNF-α production was significantly decreased by anti-Toll-like receptor (TLR)2 mAb or polymyxin B compared with the control. Furthermore, insoluble fraction-mediated cytokine production was significantly lower in peritoneal macrophages from TLR2-/- and TLR4-/- mice compared with wild-type mice. These results suggest that B. rapa L. is a potentially effective immunomodulator for activating macrophages to prevent infections.


Subject(s)
Brassica rapa/physiology , Macrophage Activation/physiology , Toll-Like Receptors/physiology , Animals , Antigens, CD/biosynthesis , Cytokines/biosynthesis , Interleukin-6/biosynthesis , Mice , Nitric Oxide/biosynthesis , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/biosynthesis
9.
Proc Natl Acad Sci U S A ; 115(33): 8418-8423, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061415

ABSTRACT

The local environment is crucial for shaping the identities of tissue-resident macrophages (Mϕs). When hemorrhage occurs in damaged tissues, hemoglobin induces differentiation of anti-inflammatory Mϕs with reparative function. Mucosal bleeding is one of the pathological features of inflammatory bowel diseases. However, the heme-mediated mechanism modulating activation of intestinal innate immune cells remains poorly understood. Here, we show that heme regulates gut homeostasis through induction of Spi-C in intestinal CX3CR1high Mϕs. Intestinal CX3CR1high Mϕs highly expressed Spi-C in a heme-dependent manner, and myeloid lineage-specific Spic-deficient (Lyz2-cre; Spicflox/flox ) mice showed severe intestinal inflammation with an increased number of Th17 cells during dextran sodium sulfate-induced colitis. Spi-C down-regulated the expression of a subset of Toll-like receptor (TLR)-inducible genes in intestinal CX3CR1high Mϕs to prevent colitis. LPS-induced production of IL-6 and IL-1α, but not IL-10 and TNF-α, by large intestinal Mϕs from Lyz2-cre; Spicflox/flox mice was markedly enhanced. The interaction of Spi-C with IRF5 was linked to disruption of the IRF5-NF-κB p65 complex formation, thereby abrogating recruitment of IRF5 and NF-κB p65 to the Il6 and Il1a promoters. Collectively, these results demonstrate that heme-mediated Spi-C is a key molecule for the noninflammatory signature of intestinal Mϕs by suppressing the induction of a subset of TLR-inducible genes through binding to IRF5.


Subject(s)
Colitis/drug therapy , Heme/pharmacology , Intestines/immunology , Macrophages/immunology , Animals , CX3C Chemokine Receptor 1/physiology , Cytokines/biosynthesis , DNA-Binding Proteins/physiology , Dextran Sulfate/toxicity , Iron, Dietary/administration & dosage , Mice , Mice, Inbred C57BL , Toll-Like Receptors/physiology , Transcription Factor RelA/physiology
10.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502383

ABSTRACT

Chemotherapy-induced intestinal mucositis, a painful debilitating condition affecting up to 40-100% of patients undergoing chemotherapy, can reduce the patients' quality of life, add health care costs and even postpone cancer treatment. In recent years, the relationships between intestinal microbiota dysbiosis and mucositis have drawn much attention in mucositis research. Chemotherapy can shape intestinal microbiota, which, in turn, can aggravate the mucositis through toll-like receptor (TLR) signaling pathways, leading to an increased expression of inflammatory mediators and elevated epithelial cell apoptosis but decreased epithelial cell differentiation and mucosal regeneration. This review summarizes relevant studies related to the relationships of mucositis with chemotherapy regimens, microbiota, TLRs, inflammatory mediators, and intestinal homeostasis, aiming to explore how gut microbiota affects the pathogenesis of mucositis and provides potential new strategies for mucositis alleviation and treatment and development of new therapies.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/microbiology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/microbiology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Drug Therapy/methods , Drug-Related Side Effects and Adverse Reactions/physiopathology , Dysbiosis/microbiology , Dysbiosis/physiopathology , Fluorouracil/pharmacology , Gastrointestinal Microbiome/physiology , Homeostasis , Humans , Intestines/microbiology , Microbiota/drug effects , Mucositis/chemically induced , Quality of Life , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Toll-Like Receptors/physiology
11.
Development ; 144(23): 4450-4461, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29097443

ABSTRACT

The transcription factor NF-κB plays an important role in the immune system, apoptosis and inflammation. Dorsal, a Drosophila homolog of NF-κB, patterns the dorsal-ventral axis in the blastoderm embryo. During this stage, Dorsal is sequestered outside the nucleus by the IκB homolog Cactus. Toll signaling on the ventral side breaks the Dorsal/Cactus complex, allowing Dorsal to enter the nucleus to regulate target genes. Fluorescent data show that Dorsal accumulates on the ventral side of the syncytial blastoderm. Here, we use modeling and experimental studies to show that this accumulation is caused by facilitated diffusion, or shuttling, of the Dorsal/Cactus complex. We also show that active Toll receptors are limiting in wild-type embryos, which is a key factor in explaining global Dorsal gradient formation. Our results suggest that shuttling is necessary for viability of embryos from mothers with compromised dorsal levels. Therefore, Cactus not only has the primary role of regulating Dorsal nuclear import, but also has a secondary role in shuttling. Given that this mechanism has been found in other, independent, systems, we suggest that it might be more prevalent than previously thought.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Models, Biological , Nuclear Proteins/physiology , Phosphoproteins/physiology , Transcription Factors/physiology , Animals , Animals, Genetically Modified , Biophysical Phenomena , Body Patterning/genetics , Body Patterning/physiology , Computer Simulation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Facilitated Diffusion , Female , Nuclear Proteins/genetics , Phosphoproteins/genetics , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/physiology , Transcription Factors/genetics
12.
Nat Immunol ; 9(4): 369-77, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18345001

ABSTRACT

Toll-like receptor (TLR) signaling is pivotal to innate and adaptive immune responses and must be tightly controlled. The mechanisms of TLR signaling have been the focus of extensive studies. Here we report that the tripartite-motif protein TRIM30alpha, a RING protein, was induced by TLR agonists and interacted with the TAB2-TAB3-TAK1 adaptor-kinase complex involved in the activation of transcription factor NF-kappaB. TRIM30alpha promoted the degradation of TAB2 and TAB3 and inhibited NF-kappaB activation induced by TLR signaling. In vivo studies showed that transfected or transgenic mice overexpressing TRIM30alpha were more resistant to endotoxic shock. Consistent with that, in vivo 'knockdown' of TRIM30alpha mRNA by small interfering RNA impaired lipopolysaccharide-induced tolerance. Finally, expression of TRIM30alpha depended on NF-kappaB activation. Our results collectively indicate that TRIM30alpha negatively regulates TLR-mediated NF-kappaB activation by targeting degradation of TAB2 and TAB3 by a 'feedback' mechanism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/physiology , NF-kappa B/metabolism , Toll-Like Receptors/physiology , Animals , Cell Line , Feedback, Physiological/immunology , Female , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/antagonists & inhibitors , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors
13.
Blood ; 132(6): 559-564, 2018 08 09.
Article in English | MEDLINE | ID: mdl-29853538

ABSTRACT

Recent studies have revealed that the intestinal bacterial microbiome plays an important role in the regulation of hematopoiesis. A correlation between adverse hematologic effects and imbalance of the intestinal microbiome, or dysbiosis, is evident in several human conditions, such as inflammatory bowel disease, obesity, and, critically, in the setting of antibiotic exposure. Here we review the effects of gut dysbiosis on the hematological compartment and our current understanding of the mechanisms through which changes in the bacterial microbiome affect hematopoiesis.


Subject(s)
Dysbiosis/complications , Gastrointestinal Microbiome , Hematopoiesis , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Bone Marrow/physiology , Dysbiosis/microbiology , Dysbiosis/physiopathology , Gastrointestinal Microbiome/drug effects , Graft Survival , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/microbiology , Mice , Models, Immunological , Myeloid Differentiation Factor 88/physiology , Neutropenia/chemically induced , Nod1 Signaling Adaptor Protein/physiology , Nutrition Disorders/complications , Nutrition Disorders/microbiology , Signal Transduction , Specific Pathogen-Free Organisms , Toll-Like Receptors/physiology
14.
Arterioscler Thromb Vasc Biol ; 39(2): 237-249, 2019 02.
Article in English | MEDLINE | ID: mdl-30567485

ABSTRACT

Objective- Perivascular adipose tissue (PVAT) is thought to play a role in vascular homeostasis and in the pathogenesis of large vessel diseases, including abdominal aortic aneurysm (AAA). Herein, we tested the hypothesis that locally restricted transcriptional profiles characterize PVAT surrounding AAA, indicating specific dysfunctions associated with the disease. Approach and Results- Using a paired sample design to limit the effects of interindividual variation, we performed a microarray-based investigation of the PVAT transcriptome in 30 patients with AAA, comparing the adipose layer of the dilated abdominal aorta with that of the not-dilated aortic neck in each patient. Furthermore, we used a state-of-the-art data mining procedure to remove the effect of confounders produced by high-throughput gene expression techniques. We found substantial differences in PVAT gene expression clearly distinguishing the dilated from the not-dilated aorta, which increased in number and magnitude with increasing AAA diameter. Comparisons with other adipose depots (omental or subcutaneous fat) confirmed that gene expression changes are locally restricted. We dissected putative mechanisms associated with AAA PVAT dysfunction through a functional enrichment network analysis: both innate and adaptive immune-response genes along with genes related to cell-death pathways, metabolic processes of collagen, sphingolipids, aminoglycans, and extracellular matrix degradation were strongly overrepresented in PVAT of AAA compared with PVAT of the not-dilated aorta. Conclusions- Our results support a possible function of PVAT in AAA pathogenesis and suggest that AAA is an immunologic disease with an underlying autoimmune component. Interfering with these disease-specific pathways would clarify their precise role in AAA pathogenesis.


Subject(s)
Adipose Tissue/immunology , Aortic Aneurysm, Abdominal/etiology , Autoimmunity , Transcriptome , Adipose Tissue/metabolism , Aged , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/metabolism , Humans , Immunity, Innate , Middle Aged , Toll-Like Receptors/physiology
15.
Nat Rev Mol Cell Biol ; 9(10): 781-95, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813294

ABSTRACT

Phagosome maturation is the process by which internalized particles (such as bacteria and apoptotic cells) are trafficked into a series of increasingly acidified membrane-bound structures, leading to particle degradation. The characterization of the phagosomal proteome and studies in model organisms and mammals have led to the identification of numerous candidate proteins that cooperate to control the maturation of phagosomes containing different particles. A subset of these candidate proteins makes up the first pathway to be identified for the maturation of apoptotic cell-containing phagosomes. This suggests that a machinery that is distinct from receptor-mediated endocytosis is used in phagosome maturation.


Subject(s)
Phagosomes/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Endocytosis/physiology , Humans , Models, Biological , Phagocytes/physiology , Phagocytes/ultrastructure , Phagocytosis/genetics , Phagocytosis/physiology , Phagosomes/genetics , Phagosomes/microbiology , Proteomics , Signal Transduction , Toll-Like Receptors/physiology , Vacuolar Proton-Translocating ATPases/physiology , rab GTP-Binding Proteins/physiology
16.
Proc Natl Acad Sci U S A ; 114(13): E2682-E2688, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28289197

ABSTRACT

The epidermis serves as a protective barrier in animals. After epidermal injury, barrier repair requires activation of many wound response genes in epidermal cells surrounding wound sites. Two such genes in Drosophila encode the enzymes dopa decarboxylase (Ddc) and tyrosine hydroxylase (ple). In this paper we explore the involvement of the Toll/NF-κB pathway in the localized activation of wound repair genes around epidermal breaks. Robust activation of wound-induced transcription from ple and Ddc requires Toll pathway components ranging from the extracellular ligand Spätzle to the Dif transcription factor. Epistasis experiments indicate a requirement for Spätzle ligand downstream of hydrogen peroxide and protease function, both of which are known activators of wound-induced transcription. The localized activation of Toll a few cell diameters from wound edges is reminiscent of local activation of Toll in early embryonic ventral hypoderm, consistent with the hypothesis that the dorsal-ventral patterning function of Toll arose from the evolutionary cooption of a morphogen-responsive function in wound repair. Furthermore, the combinatorial activity of Toll and other signaling pathways in activating epidermal barrier repair genes can help explain why developmental activation of the Toll, ERK, or JNK pathways alone fail to activate wound repair loci.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Toll-Like Receptors/physiology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation , Models, Biological , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Wound Healing/genetics
17.
COPD ; 17(6): 684-698, 2020 12.
Article in English | MEDLINE | ID: mdl-32757672

ABSTRACT

Chronic obstructive pulmonary disease is a complex pulmonary disease that causes airflow obstruction in humans. To identify the core genes in COPD pathogenesis, seven diverse microarray datasets (GSE475, GSE1122, GSE1650, GSE3212, GSE8823, GSE37768, and GSE22148) were downloaded from the gene expression omnibus database. All the datasets were analyzed independently with the R/Bioconductor package to screen the differentially expressed genes (DEGs). The gene ontology and pathway enrichment analysis were performed for the acquired DEGs using DAVID (Database for Annotation, Visualization, and Integrated Discovery). Further protein-protein interaction network was constructed for the DEGs and their potential hub genes and sub-networks were identified using Cytoscape software. From the selected seven datasets, 188 overlapped DEGs were perceived eventually based on considering the repetitive genes between at-least one dataset. Gene Ontology analysis reveals that most of the DEGs were significantly enriched in immune response, inflammatory response, extracellular region, lipid binding, cytokine, and chemokine activity. Moreover, genes from the sub-network analysis were again submitted to the DAVID server to validate the results which uncover the Toll-like receptor signaling pathway was significantly enriched and all the genes present in this pathway were likewise detected as hub genes from Cytoscape software. CXCL9, CXCL10, CXCL11, CCL4, TLR7, and SPP1 hub genes in the toll-like receptor signaling pathway were explored in this study as potential biomarker genes associated with COPD pathogenesis.


Subject(s)
Pulmonary Disease, Chronic Obstructive/etiology , Signal Transduction/physiology , Toll-Like Receptors/physiology , Biomarkers , Chemokine CCL4/genetics , Chemokines, CXC/genetics , Databases, Genetic , Gene Expression Profiling , Humans , Microarray Analysis , Osteopontin/genetics , Protein Interaction Maps
18.
Int J Mol Sci ; 21(18)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32911605

ABSTRACT

Imetelstat sodium (GRN163L; hereafter, imetelstat) is a first-in-class telomerase inhibitor that has demonstrated activity in patients with myeloproliferative neoplasms (MPNs). Treatment with imetelstat has been associated with thrombocytopenia and other hematologic adverse effects that were manageable and reversible. Toll-like receptors (TLRs) are proteins that recognize pathogen-associated molecular patterns and stimulate innate immune and pro-apoptotic responses. Because imetelstat is an oligonucleotide, and some oligonucleotides can activate TLRs, we conducted an in vitro study to rule out the possibility of imetelstat-associated thrombocytopenia by off-target effects through activation of TLRs. We used HEK293 cell lines stably co-expressing a human TLR gene and an NFκB-inducible reporter to investigate whether imetelstat can activate TLR signaling. We treated the cells with imetelstat or control oligonucleotides for 20 h, and used absorbance of the culture media to calculate the reporter activity. Treatment with imetelstat within or beyond the clinically relevant concentrations had no stimulatory effect on TLR2, TLR3, TLR4, TLR5, TLR7, or TLR9. This result was not surprising since the structure of imetelstat does not meet the reported minimal structural requirements for TLR9 activation. Furthermore, imetelstat treatment of the MPN cell line HEL did not impact the expression of TLR signaling pathway target genes that are commonly induced by activation of different TLRs, whereas it significantly reduced its target gene hTERT, human telomerase reverse transcriptase, in a dose- and time-dependent manner. Hence, cytopenias, especially thrombocytopenia observed in some patients treated with imetelstat, are not mediated by off-target interactions with TLRs.


Subject(s)
Myeloproliferative Disorders/drug therapy , Oligonucleotides/metabolism , Oligonucleotides/pharmacology , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Oligonucleotides/adverse effects , Signal Transduction/drug effects , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Thrombocytopenia/etiology , Thrombocytopenia/metabolism , Toll-Like Receptors/drug effects , Toll-Like Receptors/metabolism , Toll-Like Receptors/physiology
19.
Curr Opin Hematol ; 26(4): 207-213, 2019 07.
Article in English | MEDLINE | ID: mdl-31033704

ABSTRACT

PURPOSE OF REVIEW: The innate immune system is essential in the protection against microbial infection and facilitating tissue repair mechanisms. During these stresses, the maintenance of innate immune cell numbers through stress-induced or emergency hematopoiesis is key for our survival. One major mechanism to recognize danger signals is through the activation of Toll-like receptors (TLRs) on the surface of hematopoietic cells, including hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC), and nonhematopoietic cells, which recognize pathogen-derived or damaged-induced compounds and can influence the emergency hematopoietic response. This review explores how direct pathogen-sensing by HSC/HPC regulates hematopoiesis, and the positive and negative consequences of these signals. RECENT FINDINGS: Recent studies have highlighted new roles for TLRs in regulating HSC and HPC differentiation to innate immune cells of both myeloid and lymphoid origin and augmenting HSC and HPC migration capabilities. Most interestingly, new insights as to how acute versus chronic stimulation of TLR signaling regulates HSC and HPC function has been explored. SUMMARY: Recent evidence suggests that TLRs may play an important role in many inflammation-associated diseases. This suggests a possible use for TLR agonists or antagonists as potential therapeutics. Understanding the direct effects of TLR signaling by HSC and HPC may help regulate inflammatory/danger signal-driven emergency hematopoiesis.


Subject(s)
Hematologic Diseases/immunology , Hematopoietic Stem Cells/immunology , Inflammation/immunology , Toll-Like Receptors/physiology , Animals , Hematologic Diseases/pathology , Hematopoietic Stem Cells/cytology , Humans , Inflammation/pathology , Signal Transduction , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors
20.
Gastroenterology ; 155(5): 1539-1552.e12, 2018 11.
Article in English | MEDLINE | ID: mdl-30063920

ABSTRACT

BACKGROUND & AIMS: Activating transcription factor 6 (ATF6) regulates endoplasmic reticulum stress. We studied whether ATF6 contributes to the development of colorectal cancer (CRC) using tissue from patients and transgenic mice. METHODS: We analyzed data from 541 patients with CRC in The Cancer Genome Atlas database for genetic variants and aberrant expression levels of unfolded protein response genes. Findings were validated in a cohort of 83 patients with CRC in Germany. We generated mice with intestinal epithelial cell-specific expression of the active form of Atf6 (nATF6IEC) from 2 alleles (homozygous), mice with expression of nATF6IEC from 1 allele (heterozygous), and nATF6IECfl/fl mice (controls). All nATF6IEC mice were housed under either specific-pathogen-free or germ-free conditions. Cecal microbiota from homozygous nATF6IEC mice or control mice was transferred into homozygous nATF6IEC mice or control mice. nATF6IEC mice were crossed with mice with disruptions in the myeloid differentiation primary response gene 88 and toll-like receptor adaptor molecule 1 gene (Myd88/Trif-knockout mice). Intestinal tissues were collected from mice and analyzed by histology, immunohistochemistry, immunoblots, gene expression profiling of unfolded protein response and inflammatory genes, array-based comparative genome hybridization, and 16S ribosomal RNA gene sequencing. RESULTS: Increased expression of ATF6 was associated with reduced disease-free survival times of patients with CRC. Homozygous nATF6IEC mice developed spontaneous colon adenomas at 12 weeks of age. Compared with controls, homozygous nATF6IEC mice had changes in the profile of their cecal microbiota, increased proliferation of intestinal epithelial cells, and loss of the mucus barrier-all preceding tumor formation. These mice had increased penetration of bacteria into the inner mucus layer and activation of signal transducer and activator of transcription 3, yet inflammation was not observed at the pretumor or tumor stages. Administration of antibiotics to homozygous nATF6IEC mice greatly reduced tumor incidence, and germ-free housing completely prevented tumorigenesis. Analysis of nATF6IEC MyD88/TRIF-knockout mice showed that tumor initiation and growth required MyD88/TRIF-dependent activation of signal transducer and activator of transcription 3. Transplantation of cecal microbiota from nATF6IEC mice and control mice, collected before tumor formation, caused tumor formation in ex-germ-free nATF6IEC mice. CONCLUSIONS: In patients with CRC, ATF6 was associated with reduced time of disease-free survival. In studies of nATF6IEC mice, we found sustained intestinal activation of ATF6 in the colon to promote dysbiosis and microbiota-dependent tumorigenesis.


Subject(s)
Activating Transcription Factor 6/physiology , Colorectal Neoplasms/etiology , Dysbiosis/etiology , Immunity, Innate , Intestines/microbiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Colorectal Neoplasms/mortality , Disease Progression , Humans , Mice , Myeloid Differentiation Factor 88/physiology , STAT3 Transcription Factor/physiology , Toll-Like Receptors/physiology , Unfolded Protein Response
SELECTION OF CITATIONS
SEARCH DETAIL