Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31964750

ABSTRACT

Human genital Chlamydia infection is a major public health concern due to the serious reproductive system complications. Chlamydia binds several receptor tyrosine kinases (RTKs) on host cells, including the epidermal growth factor receptor (EGFR), and activates cellular signaling cascades for host invasion, cytoskeletal remodeling, optimal inclusion development, and induction of pathogenic epithelial-mesenchyme transition (EMT). Chlamydia also upregulates transforming growth factor beta (TGF-ß) expression, whose signaling pathway synergizes with the EGFR cascade, but its role in infectivity, inclusions, and EMT induction is unknown. We hypothesized that the EGFR and TGF-ß signaling pathways cooperate during chlamydial infection for optimal inclusion development and stable EMT induction. The results revealed that Chlamydia upregulated TGF-ß expression as early as 6 h postinfection of epithelial cells and stimulated both the EGFR and TGF-ß signaling pathways. Inhibition of either the EGFR or TGF-ßR1 signaling substantially reduced inclusion development; however, the combined inhibition of both EGFR and TGF-ßR1 signaling reduced inclusions by over 90% and prevented EMT induction. Importantly, EGFR inhibition suppressed TGF-ß expression, and an inhibitory thrombospondin-1 (Tsp1)-based peptide inhibited chlamydia-induced EMT, revealing a major source of active TGF-ß during infection. Finally, TGF-ßR signaling inhibition suppressed the expression of transforming acidic coiled-coil protein-3 (TACC3), which stabilizes EGFR signaling, suggesting reciprocal regulation between TGF-ß and EGFR signaling during chlamydial infection. Thus, RTK-mediated host invasion by chlamydia upregulated TGF-ß expression and signaling, which cooperated with other cellular signaling cascades and cytoskeletal remodeling to support optimal inclusion development and EMT induction. This finding may provide new targets for chlamydial disease biomarkers and prevention.


Subject(s)
Chlamydia Infections/physiopathology , Chlamydia/growth & development , Epithelial Cells/microbiology , ErbB Receptors/metabolism , Host-Pathogen Interactions , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Endocytosis , Epithelial-Mesenchymal Transition , Inclusion Bodies/microbiology , Mice , Models, Biological
2.
Biochem Biophys Res Commun ; 508(2): 421-429, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30503337

ABSTRACT

The unfolded protein response (UPR) contributes to chlamydial pathogenesis, as a source of lipids and ATP during replication, and for establishing the initial anti-apoptotic state of host cell that ensures successful inclusion development. The molecular mechanism(s) of UPR induction by Chlamydia is unknown. Chlamydia use type III secretion system (T3SS) effector proteins (e.g, the Translocated Actin-Recruiting Phosphoprotein (Tarp) to stimulate host cell's cytoskeletal reorganization that facilitates invasion and inclusion development. We investigated the hypothesis that T3SS effector-mediated assembly of myosin-II complex produces activated non-muscle myosin heavy chain II (NMMHC-II), which then binds the UPR master regulator (BiP) and/or transducers to induce UPR. Our results revealed the interaction of the chlamydial effector proteins (CT228 and Tarp) with components of the myosin II complex and UPR regulator and transducer during infection. These interactions caused the activation and binding of NMMHC-II to BiP and IRE1α leading to UPR induction. In addition, specific inhibitors of myosin light chain kinase, Tarp oligomerization and myosin ATPase significantly reduced UPR activation and Chlamydia replication. Thus, Chlamydia induce UPR through T3SS effector-mediated activation of NMMHC-II components of the myosin complex to facilitate infectivity. The finding provides greater insights into chlamydial pathogenesis with the potential to identify therapeutic targets and formulations.


Subject(s)
Chlamydia muridarum/pathogenicity , Chlamydia trachomatis/pathogenicity , Host Microbial Interactions/physiology , Unfolded Protein Response/physiology , Animals , Chlamydia Infections/etiology , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Chlamydia muridarum/metabolism , Chlamydia trachomatis/metabolism , HeLa Cells , Humans , Inclusion Bodies/metabolism , Mice , Myosin Type II/metabolism , Type III Secretion Systems/metabolism
3.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29084894

ABSTRACT

The reproductive system complications of genital chlamydial infection include fallopian tube fibrosis and tubal factor infertility. However, the molecular pathogenesis of these complications remains poorly understood. The induction of pathogenic epithelial-mesenchymal transition (EMT) through microRNA (miRNA) dysregulation was recently proposed as the pathogenic basis of chlamydial complications. Focusing on fibrogenesis, we investigated the hypothesis that chlamydia-induced fibrosis is caused by EMT-driven generation of myofibroblasts, the effector cells of fibrosis that produce excessive extracellular matrix (ECM) proteins. The results revealed that the targets of a major category of altered miRNAs during chlamydial infection are key components of the pathophysiological process of fibrogenesis; these target molecules include collagen types I, III, and IV, transforming growth factor ß (TGF-ß), TGF-ß receptor 1 (TGF-ßR1), connective tissue growth factor (CTGF), E-cadherin, SRY-box 7 (SOX7), and NFAT (nuclear factor of activated T cells) kinase dual-specificity tyrosine (Y) phosphorylation-regulated kinase 1a (Dyrk1a). Chlamydial induction of EMT resulted in the generation of α-smooth muscle actin (α-SMA)-positive myofibroblasts that produced ECM proteins, including collagen types I and III and fibronectin. Furthermore, the inhibition of EMT prevented the generation of myofibroblasts and production of ECM proteins during chlamydial infection. These findings may provide useful avenues for targeting EMT or specific components of the EMT pathways as a therapeutic intervention strategy to prevent chlamydia-related complications.


Subject(s)
Chlamydia Infections/complications , Chlamydia Infections/pathology , Chlamydia/pathogenicity , Epithelial-Mesenchymal Transition/physiology , Fibrosis/etiology , Fibrosis/pathology , Actins/metabolism , Animals , Cadherins/metabolism , Cell Line , Chlamydia Infections/microbiology , Collagen/metabolism , Connective Tissue Growth Factor/metabolism , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Fibrosis/microbiology , Mice , MicroRNAs/metabolism , Myofibroblasts/microbiology , Myofibroblasts/pathology , NFATC Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , SOXF Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
4.
J Infect Dis ; 215(3): 456-465, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27932618

ABSTRACT

Chlamydia is an obligate intracellular bacterium that relies on host cells for essential nutrients and adenosine triphosphate (ATP) for a productive infection. Although the unfolded protein response (UPR) plays a major role in certain microbial infectivity, its role in chlamydial pathogenesis is unknown. We hypothesized that Chlamydia induces UPR and exploits it to upregulate host cell uptake and metabolism of glucose, production of ATP, phospholipids, and other molecules required for its replicative development and host survival. Using a combination of biochemical and pathway inhibition assays, we showed that the 3 UPR pathway transducers-protein kinase RNA-activated (PKR)-like ER kinase (PERK), inositol-requiring enzyme-1α (IRE1α), and activating transcription factor-6α (ATF6α)-were activated during Chlamydia infection. The kinase activity of PERK and ribonuclease (RNase) of IRE1α mediated the upregulation of hexokinase II and production of ATP via substrate-level phosphorylation. In addition, the activation of PERK and IRE1α promoted autophagy formation and apoptosis resistance for host survival. Moreover, the activation of IRE1α resulted in the generation of spliced X-box binding protein 1 (sXBP1) and upregulation of lipid production. The vital role of UPR pathways in Chlamydia development and pathogenesis could lead to the identification of potential molecular targets for therapeutics against Chlamydia.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia/pathogenicity , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Animals , Apoptosis , Cell Survival , Chlamydia Infections/metabolism , Endoribonucleases/metabolism , Enzyme Activation , Female , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , eIF-2 Kinase/metabolism
5.
PLoS One ; 10(12): e0145198, 2015.
Article in English | MEDLINE | ID: mdl-26681200

ABSTRACT

Chlamydia trachomatis genital infection in women causes serious adverse reproductive complications, and is a strong co-factor for human papilloma virus (HPV)-associated cervical epithelial carcinoma. We tested the hypothesis that Chlamydia induces epithelial-mesenchyme transition (EMT) involving T cell-derived TNF-alpha signaling, caspase activation, cleavage inactivation of dicer and dysregulation of micro-RNA (miRNA) in the reproductive epithelium; the pathologic process of EMT causes fibrosis and fertility-related epithelial dysfunction, and also provides the co-factor function for HPV-related cervical epithelial carcinoma. Using a combination of microarrays, immunohistochemistry and proteomics, we showed that chlamydia altered the expression of crucial miRNAs that control EMT, fibrosis and tumorigenesis; specifically, miR-15a, miR-29b, miR-382 and MiR-429 that maintain epithelial integrity were down-regulated, while miR-9, mi-R-19a, miR-22 and miR-205 that promote EMT, fibrosis and tumorigenesis were up-regulated. Chlamydia induced EMT in vitro and in vivo, marked by the suppression of normal epithelial cell markers especially E-cadherin but up-regulation of mesenchymal markers of pathological EMT, including T-cadherin, MMP9, and fibronectin. Also, Chlamydia upregulated pro-EMT regulators, including the zinc finger E-box binding homeobox protein, ZEB1, Snail1/2, and thrombospondin1 (Thbs1), but down-regulated anti-EMT and fertility promoting proteins (i.e., the major gap junction protein connexin 43 (Cx43), Mets1, Add1Scarb1 and MARCKSL1). T cell-derived TNF-alpha signaling was required for chlamydial-induced infertility and caspase inhibitors prevented both infertility and EMT. Thus, chlamydial-induced T cell-derived TNF-alpha activated caspases that inactivated dicer, causing alteration in the expression of reproductive epithelial miRNAs and induction of EMT. EMT causes epithelial malfunction, fibrosis, infertility, and the enhancement of tumorigenesis of HPV oncogene-transformed epithelial cells. These findings provide a novel understanding of the molecular pathogenesis of chlamydia-associated diseases, which may guide a rational prevention strategy.


Subject(s)
Chlamydia Infections/metabolism , Epithelial-Mesenchymal Transition , Animals , Cadherins/genetics , Cadherins/metabolism , Caspases/metabolism , Chlamydia Infections/pathology , Female , Fibronectins/genetics , Fibronectins/metabolism , HeLa Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Snail Family Transcription Factors , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Zinc Finger E-box-Binding Homeobox 1
6.
J Infect Dis ; 207(7): 1095-104, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303804

ABSTRACT

Tubal factor infertility (TFI) represents 36% of female infertility and genital infection by Chlamydia trachomatis (C. trachomatis) is a major cause. Although TFI is associated with host inflammatory responses to bacterial components, the molecular pathogenesis of Chlamydia-induced infertility remains poorly understood. We investigated the hypothesis that activation of specific cysteine proteases, the caspases, during C. trachomatis genital infection causes the disruption of key fertility-promoting molecules required for embryo development and implantation. We analyzed the effect of caspase inhibition on infertility and the integrity of Dicer, a caspase-sensitive, fertility-promoting ribonuclease III enzyme, and key micro-RNAs in the reproductive system. Genital infection with the inflammation- and caspase-inducing, wild-type C. trachomatis serovar L2 led to infertility, but the noninflammation-inducing, plasmid-free strain did not. We confirmed that caspase-mediated apoptotic tissue destruction may contribute to chlamydial pathogenesis. Caspase-1 or -3 deficiency, or local administration of the pan caspase inhibitor, Z-VAD-FMK into normal mice protected against Chlamydia-induced infertility. Finally, the oviducts of infected infertile mice showed evidence of caspase-mediated cleavage inactivation of Dicer and alteration in critical miRNAs that regulate growth, differentiation, and development, including mir-21. These results provide new insight into the molecular pathogenesis of TFI with significant implications for new strategies for treatment and prevention of chlamydial complications.


Subject(s)
Caspase 1/metabolism , Caspase 3/metabolism , Chlamydia trachomatis/pathogenicity , Infertility, Female/microbiology , Infertility, Female/prevention & control , Pregnancy Complications, Infectious/prevention & control , Animals , Apoptosis , Caspase 1/genetics , Caspase 3/genetics , Chlamydia Infections/enzymology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Enzyme Activation , Female , HeLa Cells , Humans , Infertility, Female/enzymology , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Pregnancy Complications, Infectious/enzymology , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/pathology
7.
J Immunoassay Immunochem ; 31(3): 250-7, 2010.
Article in English | MEDLINE | ID: mdl-20623410

ABSTRACT

A comparative analysis was performed to determine the sensitivity and efficiency of three fluorescent labeling techniques, including direct fluorescent-antibody staining (FA), Zenon labeling, and quantum dot (QD) nanocrystal technology. Two varicella-zoster virus immunoglobin (Ig) G forms, mAb 4F9 and mAb g62, were selected for these studies. The results indicated that: (1) All three methods demonstrated similar brightness and photostability; (2) the time required to conjugate the antibody varied, with Zenon labeling being the quickest; and (3) the stability of each conjugated complex was different, with FITC/rhodamine-conjugated antibody being the most stable.


Subject(s)
Fluorescence , Fluorescent Antibody Technique/methods , Nanostructures/chemistry , Nanotechnology/methods , Quantum Dots , Staining and Labeling/methods , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Fluorescent Dyes/chemistry , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Isothiocyanates/chemistry , Rhodamines/chemistry
8.
Prostate ; 68(3): 287-95, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18163430

ABSTRACT

BACKGROUND: A signaling interaction between transforming growth factor-beta (TGF-beta) and androgens promotes apoptosis in human prostate cancer cells LNCaP-TbetaRII (androgen-sensitive and TGF-beta responsive). This study investigated the contribution of androgen receptor (AR) in the combined effect of TGF-beta and dihydrotestosterone (DHT), on regulation of apoptosis and AR- and TGF-beta mediated transcriptional activity in human prostate cancer cells. METHODS: Transcriptional activation in response to TGF-beta (5 ng/ml) and DHT (1 nM) was evaluated using transient transfections and luciferase assays in human prostate cancer cells, LNCaP-TbetaRII and PC-3, overexpressing the wild type AR. The apoptotic response to DHT/TGFbeta treatment was correlated with AR cellular distribution and the AR interaction with TGF-beta intracellular effector Smad4. RESULTS: The results revealed that TGF-beta signaling induced AR-mediated transcriptional activation in two androgen-responsive promoters [probasin and prostate specific antigen (PSA)]. TGF-beta1 induced transcriptional activity enhanced by DHT in both cell lines (LNCaP-TbetaRII and PC-3-AR) via AR-Smad4 interaction. This interaction however does not exclusively drive TGF-beta mediated apoptosis as DHT failed to enhance such an effect in PC-3 AR (wt) cells. CONCLUSIONS: These results demonstrate that the AR status determines the sensitivity of prostate cancer cells to the apoptotic effects of TGF-beta1, thus providing a new insight into the mechanism via which TGF-beta cross-sections the AR axis toward the functional convergence of the two pathways in the development of androgen-independent prostate cancer. This study is potentially significant in defining the contribution of AR status to the emergence of androgen-independent prostate tumors.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Transforming Growth Factor beta/metabolism , Androgen-Binding Protein/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Drug Synergism , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Microscopy, Fluorescence , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad4 Protein/metabolism , Transcriptional Activation/drug effects , Transfection , Transforming Growth Factor beta/pharmacology
9.
J Neurosci Res ; 70(6): 784-93, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12444600

ABSTRACT

Target ablation by olfactory bulbectomy synchronizes the degenerative cell death of olfactory receptor neurons (ORNs), infiltration of macrophages, and proliferation of progenitor cells, leading to neurogenesis, ORN replacement, and regeneration of the sensory epithelium. Although macrophages participate in the degenerative and regenerative events, little is known of the molecular and cellular mechanisms associated with their recruitment during the earliest period following target ablation. Macrophage inflammatory protein-1alpha (MIP-1alpha) and monocyte chemoattractant protein-1 (MCP-1), which are members of the CC or beta-chemokine subfamily, are chemoattractants for monocytes/macrophages. Shortly after target ablation, the protein and mRNA levels for MIP-1alpha and MCP-1 were up-regulated, showing peak expression levels from 16 hr to 3 days post-OBX; this coincided with the pattern of infiltration of activated F4/80(+) macrophages. The mRNAs for MIP-1alpha and MCP-1, as well as their cognate receptors CCR1 and CCR2, respectively, were localized in resident and infiltrating macrophages in numbers commensurate with those of F4/80-immunopositive macrophages in adjacent tissue sections. The mRNA(+) macrophages were localized within olfactory epithelial compartments that corresponded with their proposed functions associated with phagocytosis, proliferation, and infiltration. Our data support the hypothesis that MIP-1alpha and MCP-1 are chemoattractant chemokines associated with the recruitment of macrophages into the olfactory epithelium shortly after target ablation.


Subject(s)
Chemokine CCL2/biosynthesis , Macrophage Inflammatory Proteins/biosynthesis , Macrophages/cytology , Olfactory Mucosa/cytology , RNA, Messenger/analysis , Animals , Axotomy , Cell Movement/physiology , Chemokine CCL2/genetics , Chemokine CCL3 , Chemokine CCL4 , Chemotactic Factors , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , In Situ Hybridization , Macrophage Inflammatory Proteins/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Olfactory Bulb/physiology , Olfactory Mucosa/innervation , Olfactory Mucosa/metabolism , Polymerase Chain Reaction , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Time Factors , Up-Regulation
10.
J Neurosci Res ; 67(2): 246-54, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11782968

ABSTRACT

After target ablation by olfactory bulbectomy (OBX), the murine olfactory epithelium (OE) undergoes degenerative changes leading to apoptosis of olfactory receptor neurons (ORNs) followed by regenerative changes that include proliferation of progenitor cells leading to neurogenesis and ORN replacement. Macrophages recruited to the OE after OBX are involved in both the degenerative and regenerative processes. Relative quantitative RT-PCR was used to demonstrate that within hours of OBX, mRNAs encoding three key components in the leukemia inhibitory factor (LIF) signaling pathway, including LIF, LIF receptor (LIFR), and STAT3, as well as cyclin D1, a growth factor sensor indicative of progenitor cell transformation, were upregulated. These mRNAs reached peak levels of expression on or before day 3 post-OBX, coincident with the peak time for macrophage recruitment and progenitor cell proliferation. Cells expressing LIF mRNA in the OE of mice at 3 days post-OBX, the time point at which LIF mRNA expression peaked, were identified using non-isotopic in situ hybridization. LIF mRNA was localized in infiltrating macrophages; near-adjacent sections exhibited macrophages immunoreactive for F4/80, a marker for activated macrophages, in numbers commensurate with those expressing LIF mRNA. LIF mRNA was also localized in surviving ORNs, identified by their expression of olfactory marker protein (OMP) mRNA and protein in near-adjacent sections. Our data suggest that LIF functions as a mitogen originating from recruited macrophages through an intercellular signaling pathway that stimulates proliferation of progenitor cells leading to neurogenesis and regeneration, and as an intracellular survival factor for traumatized ORNs.


Subject(s)
Growth Inhibitors/genetics , Interleukin-6 , Lymphokines/genetics , Macrophages/metabolism , Nerve Regeneration/genetics , Olfactory Receptor Neurons/metabolism , Retrograde Degeneration/metabolism , Stem Cells/metabolism , Up-Regulation/genetics , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Axotomy , Cell Movement/physiology , Cyclin D1/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Immunohistochemistry , Leukemia Inhibitory Factor , Leukemia Inhibitory Factor Receptor alpha Subunit , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Olfactory Bulb/injuries , Olfactory Bulb/surgery , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/cytology , Phenotype , RNA, Messenger/metabolism , Receptors, Cytokine/genetics , Receptors, OSM-LIF , Retrograde Degeneration/physiopathology , STAT3 Transcription Factor , Signal Transduction/genetics , Stem Cells/cytology , Time Factors , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...