Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cell Rep ; 43(5): 114156, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38687642

ABSTRACT

The maintenance of antigen-specific CD8+ T cells underlies the efficacy of vaccines and immunotherapies. Pathways contributing to CD8+ T cell loss are not completely understood. Uncovering the pathways underlying the limited persistence of CD8+ T cells would be of significant benefit for developing novel strategies of promoting T cell persistence. Here, we demonstrate that murine CD8+ T cells experience endoplasmic reticulum (ER) stress following activation and that the ER-associated degradation (ERAD) adapter Sel1L is induced in activated CD8+ T cells. Sel1L loss limits CD8+ T cell function and memory formation following acute viral infection. Mechanistically, Sel1L is required for optimal bioenergetics and c-Myc expression. Finally, we demonstrate that human CD8+ T cells experience ER stress upon activation and that ER stress is negatively associated with improved T cell functionality in T cell-redirecting therapies. Together, these results demonstrate that ER stress and ERAD are important regulators of T cell function and persistence.


Subject(s)
CD8-Positive T-Lymphocytes , Endoplasmic Reticulum Stress , Endoplasmic Reticulum-Associated Degradation , Immunologic Memory , Animals , Humans , Mice , Acute Disease , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Intracellular Signaling Peptides and Proteins , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic Choriomeningitis/pathology , Mice, Inbred C57BL , Proteins , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Male , Female
2.
Proc Natl Acad Sci U S A ; 121(17): e2318420121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38621136

ABSTRACT

In response to an immune challenge, naive T cells undergo a transition from a quiescent to an activated state acquiring the effector function. Concurrently, these T cells reprogram cellular metabolism, which is regulated by iron. We and others have shown that iron homeostasis controls proliferation and mitochondrial function, but the underlying mechanisms are poorly understood. Given that iron derived from heme makes up a large portion of the cellular iron pool, we investigated iron homeostasis in T cells using mice with a T cell-specific deletion of the heme exporter, FLVCR1 [referred to as knockout (KO)]. Our finding revealed that maintaining heme and iron homeostasis is essential to keep naive T cells in a quiescent state. KO naive CD4 T cells exhibited an iron-overloaded phenotype, with increased spontaneous proliferation and hyperactive mitochondria. This was evidenced by reduced IL-7R and IL-15R levels but increased CD5 and Nur77 expression. Upon activation, however, KO CD4 T cells have defects in proliferation, IL-2 production, and mitochondrial functions. Iron-overloaded CD4 T cells failed to induce mitochondrial iron and exhibited more fragmented mitochondria after activation, making them susceptible to ferroptosis. Iron overload also led to inefficient glycolysis and glutaminolysis but heightened activity in the hexosamine biosynthetic pathway. Overall, these findings highlight the essential role of iron in controlling mitochondrial function and cellular metabolism in naive CD4 T cells, critical for maintaining their quiescent state.


Subject(s)
CD4-Positive T-Lymphocytes , Iron , Mice , Animals , Iron/metabolism , Mitochondria/metabolism , Signal Transduction , Heme/metabolism
3.
Science ; 383(6688): eadk6176, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38484056

ABSTRACT

Obeldesivir (ODV, GS-5245) is an orally administered prodrug of the parent nucleoside of remdesivir (RDV) and is presently in phase 3 trials for COVID-19 treatment. In this work, we show that ODV and its circulating parent nucleoside metabolite, GS-441524, have similar in vitro antiviral activity against filoviruses, including Marburg virus, Ebola virus, and Sudan virus (SUDV). We also report that once-daily oral ODV treatment of cynomolgus monkeys for 10 days beginning 24 hours after SUDV exposure confers 100% protection against lethal infection. Transcriptomics data show that ODV treatment delayed the onset of inflammation and correlated with antigen presentation and lymphocyte activation. Our results offer promise for the further development of ODV to control outbreaks of filovirus disease more rapidly.


Subject(s)
Alanine , Antiviral Agents , Ebolavirus , Hemorrhagic Fever, Ebola , Nucleosides , Prodrugs , Animals , Administration, Oral , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Macaca fascicularis , Nucleosides/administration & dosage , Nucleosides/pharmacology , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/pharmacology , Alanine/administration & dosage , Alanine/analogs & derivatives , Alanine/pharmacology , Prodrugs/administration & dosage , Prodrugs/pharmacology , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology
4.
bioRxiv ; 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38328232

ABSTRACT

Photosensitivity is observed in numerous autoimmune diseases and drives poor quality of life and disease flares. Elevated epidermal type I interferon (IFN) production primes for photosensitivity and enhanced inflammation, but the substrates that sustain and amplify this cycle remain undefined. Here, we show that IFN-induced Z-DNA binding protein 1 (ZBP1) stabilizes ultraviolet (UV)B-induced cytosolic Z-DNA derived from oxidized mitochondrial DNA. ZBP1 is significantly upregulated in the epidermis of adult and pediatric patients with autoimmune photosensitivity. Strikingly, lupus keratinocytes accumulate extensive cytosolic Z-DNA after UVB, and transfection of keratinocytes with Z-DNA results in stronger IFN production through cGAS-STING activation compared to B-DNA. ZBP1 knockdown abrogates UV-induced IFN responses, whereas overexpression results in a lupus-like phenotype with spontaneous Z-DNA accumulation and IFN production. Our results highlight Z-DNA and ZBP1 as critical mediators for UVB-induced inflammation and uncover how type I IFNs prime for cutaneous inflammation in photosensitivity.

5.
Emerg Microbes Infect ; 13(1): 2301061, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38164768

ABSTRACT

Lassa virus (LASV) is a World Health Organization (WHO) priority pathogen that causes high morbidity and mortality. Recently, we showed that a combination of three broadly neutralizing human monoclonal antibodies known as Arevirumab-3 (8.9F, 12.1F, 37.2D) based on the lineage IV Josiah strain protected 100% of cynomolgus macaques against heterologous challenge with lineage II and III strains of LASV when therapy was initiated beginning at day 8 after challenge. LASV strains from Benin and Togo represent a new lineage VII that are more genetically diverse from lineage IV than strains from lineages II and III. Here, we tested the ability of Arevirumab-3 to protect macaques against a LASV lineage VII Togo isolate when treatment was administered beginning 8 days after exposure. Unexpectedly, only 40% of treated animals survived challenge. In a subsequent study we showed that Arevirumab-3 protected 100% of macaques from lethal challenge when treatment was initiated 7 days after LASV Togo exposure. Based on our transcriptomics data, successful Arevirumab-3 treatment correlated with diminished neutrophil signatures and the predicted development of T cell responses. As the in vitro antiviral activity of Arevirumab-3 against LASV Togo was equivalent to lineage II and III strains, the reduced protection in macaques against Togo likely reflects the faster disease course of LASV Togo in macaques than other strains. This data causes concern regarding the ability of heterologous vaccines and treatments to provide cross protection against lineage VII LASV isolates.


Subject(s)
Lassa Fever , Lassa virus , Humans , Animals , Virulence , Macaca fascicularis , Antibodies, Monoclonal/pharmacology
6.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873171

ABSTRACT

Infection by intracellular pathogens can trigger activation of the IRE1α branch of the unfolded protein response (UPR), which then modulates innate immunity and infection outcomes during bacterial or viral infection. However, the mechanisms by which infection activates IRE1α have not been fully elucidated. While recognition of microbe-associated molecular patterns can activate IRE1α, it is unclear whether this depends on the canonical role of IRE1α in detecting misfolded proteins. Here, we report that Candida albicans infection of macrophages results in IRE1α activation through C-type lectin receptor signaling, reinforcing a role for IRE1α as a central regulator of host responses to infection by a broad range of pathogens. However, IRE1α activation was not preceded by protein misfolding in response to either C. albicans infection or lipopolysaccharide treatment, implicating a non-canonical mode of IRE1α activation after recognition of microbial patterns. Investigation of the phenotypic consequences of IRE1α activation in macrophage antimicrobial responses revealed that IRE1α activity enhances the fungicidal activity of macrophages. Macrophages lacking IRE1α activity displayed inefficient phagolysosomal fusion, enabling C. albicans to evade fungal killing and escape the phagosome. Together, these data provide mechanistic insight for the non-canonical activation of IRE1α during infection, and reveal central roles for IRE1α in macrophage antifungal responses.

7.
J Infect Dis ; 228(Suppl 7): S660-S670, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37171813

ABSTRACT

BACKGROUND: The family Filoviridae consists of several virus members known to cause significant mortality and disease in humans. Among these, Ebola virus (EBOV), Marburg virus (MARV), Sudan virus (SUDV), and Bundibugyo virus (BDBV) are considered the deadliest. The vaccine, Ervebo, was shown to rapidly protect humans against Ebola disease, but is indicated only for EBOV infections with limited cross-protection against other filoviruses. Whether multivalent formulations of similar recombinant vesicular stomatitis virus (rVSV)-based vaccines could likewise confer rapid protection is unclear. METHODS: Here, we tested the ability of an attenuated, quadrivalent panfilovirus VesiculoVax vaccine (rVSV-Filo) to elicit fast-acting protection against MARV, EBOV, SUDV, and BDBV. Groups of cynomolgus monkeys were vaccinated 7 days before exposure to each of the 4 viral pathogens. All subjects (100%) immunized 1 week earlier survived MARV, SUDV, and BDBV challenge; 80% survived EBOV challenge. Survival correlated with lower viral load, higher glycoprotein-specific immunoglobulin G titers, and the expression of B-cell-, cytotoxic cell-, and antigen presentation-associated transcripts. CONCLUSIONS: These results demonstrate multivalent VesiculoVax vaccines are suitable for filovirus outbreak management. The highly attenuated nature of the rVSV-Filo vaccine may be preferable to the Ervebo "delta G" platform, which induced adverse events in a subset of recipients.


Subject(s)
Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Marburgvirus , Viral Vaccines , Humans , Animals , Vaccines, Attenuated , Macaca fascicularis , Vesiculovirus/genetics , Vesicular stomatitis Indiana virus , Antibodies, Viral
8.
Sci Adv ; 9(5): eade8701, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36735777

ABSTRACT

Macrophage metabolic plasticity enables repurposing of electron transport from energy generation to inflammation and host defense. Altered respiratory complex II function has been implicated in cancer, diabetes, and inflammation, but regulatory mechanisms are incompletely understood. Here, we show that macrophage inflammatory activation triggers Complex II disassembly and succinate dehydrogenase subunit B loss through sequestration and selective mitophagy. Mitochondrial fission supported lipopolysaccharide-stimulated succinate dehydrogenase subunit B degradation but not sequestration. We hypothesized that this Complex II regulatory mechanism might be coordinated by the mitochondrial phospholipid cardiolipin. Cardiolipin synthase knockdown prevented lipopolysaccharide-induced metabolic remodeling and Complex II disassembly, sequestration, and degradation. Cardiolipin-depleted macrophages were defective in lipopolysaccharide-induced pro-inflammatory cytokine production, a phenotype partially rescued by Complex II inhibition. Thus, cardiolipin acts as a critical organizer of inflammatory metabolic remodeling.


Subject(s)
Cardiolipins , Succinate Dehydrogenase , Humans , Succinate Dehydrogenase/metabolism , Cardiolipins/metabolism , Lipopolysaccharides/pharmacology , Mitochondria/metabolism , Inflammation/metabolism
9.
J Cell Sci ; 135(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35107133

ABSTRACT

The internalization of solutes by macropinocytosis provides an essential route for nutrient uptake in many cells. Macrophages increase macropinocytosis in response to growth factors and other stimuli. To test the hypothesis that nutrient environments modulate solute uptake by macropinocytosis, this study analyzed the effects of extracellular amino acids on the accumulation of fluorescent fluid-phase probes in murine macrophages. Nine amino acids, added individually or together, were capable of suppressing macropinocytosis in murine bone marrow-derived macrophages stimulated with the growth factors colony stimulating factor 1 (CSF1) or interleukin 34, both ligands of the CSF1 receptor (CSF1R). The suppressive amino acids did not inhibit macropinocytosis in response to lipopolysaccharide, the chemokine CXCL12, or the tumor promoter phorbol myristate acetate. Suppressive amino acids promoted release of CSF1R from cells and resulted in the formation of smaller macropinosomes in response to CSF1. This suppression of growth factor-stimulated macropinocytosis indicates that different nutrient environments modulate CSF1R levels and bulk ingestion by macropinocytosis, with likely consequences for macrophage growth and function.


Subject(s)
Amino Acids , Macrophage Colony-Stimulating Factor , Animals , Endosomes/metabolism , Macrophages/metabolism , Mice , Pinocytosis/drug effects , Receptor Protein-Tyrosine Kinases/metabolism
10.
J Immunol ; 205(10): 2806-2820, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33055281

ABSTRACT

Intermediate monocytes (iMo; CD14+CD16+) increase in number in the circulation of patients with unstable coronary artery disease (CAD), and their recruitment to inflamed arteries is implicated in events leading to mortality following MI. Monocyte recruitment to inflamed coronary arteries is initiated by high affinity ß2-integrin (CD11c/CD18) that activates ß1-integrin (VLA-4) to bind endothelial VCAM-1. How integrin binding under shear stress mechanosignals a functional shift in iMo toward an inflammatory phenotype associated with CAD progression is unknown. Whole blood samples from patients treated for symptomatic CAD including non-ST elevation MI, along with healthy age-matched subjects, were collected to assess chemokine and integrin receptor levels on monocytes. Recruitment on inflamed human aortic endothelium or rVCAM-1 under fluid shear stress was assessed using a microfluidic-based artery on a chip (A-Chip). Membrane upregulation of high affinity CD11c correlated with concomitant activation of VLA-4 within focal adhesive contacts was required for arrest and diapedesis across inflamed arterial endothelium to a greater extent in non-ST elevation MI compared with stable CAD patients. The subsequent conversion of CD11c from a high to low affinity state under fluid shear activated phospho-Syk- and ADAM17-mediated proteolytic cleavage of CD16. This marked the conversion of iMo to an inflammatory phenotype associated with nuclear translocation of NF-κB and production of IL-1ß+ We conclude that CD11c functions as a mechanoregulator that activates an inflammatory state preferentially in a majority of iMo from cardiac patients but not healthy patients.


Subject(s)
CD11c Antigen/metabolism , Coronary Artery Disease/immunology , Endothelium, Vascular/immunology , Monocytes/immunology , Non-ST Elevated Myocardial Infarction/immunology , Adult , Aged , Allosteric Regulation/immunology , Aorta/cytology , Case-Control Studies , Cell Culture Techniques , Cell Line , Cell Membrane/metabolism , Coronary Artery Disease/blood , Coronary Artery Disease/surgery , Coronary Vessels/cytology , Coronary Vessels/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Integrin alpha4beta1/metabolism , Lab-On-A-Chip Devices , Male , Microfluidic Analytical Techniques/instrumentation , Middle Aged , Non-ST Elevated Myocardial Infarction/blood , Non-ST Elevated Myocardial Infarction/surgery , Percutaneous Coronary Intervention , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Transendothelial and Transepithelial Migration/immunology , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
11.
Front Cell Infect Microbiol ; 10: 593805, 2020.
Article in English | MEDLINE | ID: mdl-33520735

ABSTRACT

The mitochondrial network plays a critical role in the regulation of innate immune signaling and subsequent production of proinflammatory cytokines such as IFN-ß and IL-1ß. Dynamin-related protein 1 (DRP1) promotes mitochondrial fission and quality control to maintain cellular homeostasis during infection. However, mechanisms by which DRP1 and mitochondrial dynamics control innate immune signaling and the proinflammatory response are incompletely understood. Here we show that macrophage DRP1 is a positive regulator of TNF-α production during sterile inflammation or bacterial infection. Silencing macrophage DRP1 decreased mitochondrial fragmentation and TNF-α production upon stimulation with lipopolysaccharide (LPS) or methicillin-resistant Staphylococcus aureus (MRSA) infection. The defect in TNF-α induction could not be attributed to changes in gene expression. Instead, DRP1 was required for post-transcriptional control of TNF-α. In contrast, silencing DRP1 enhanced IL-6 and IL-1ß production, indicating a distinct mechanism for DRP1-dependent TNF-α regulation. Our results highlight DRP1 as a key player in the macrophage pro-inflammatory response and point to its involvement in post-transcriptional control of TNF-α production.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Mitochondrial Dynamics , Dynamins , Mitochondria , Mitochondrial Proteins/genetics , Tumor Necrosis Factor-alpha
13.
J Vis Exp ; (144)2019 02 28.
Article in English | MEDLINE | ID: mdl-30882782

ABSTRACT

Staphylococcus aureus (S. aureus) infections, including methicillin resistant stains, are an enormous burden on the healthcare system. With incidence rates of S. aureus infection climbing annually, there is a demand for additional research in its pathogenicity. Animal models of infectious disease advance our understanding of the host-pathogen response and lead to the development of effective therapeutics. Neutrophils play a primary role in the innate immune response that controls S. aureus infections by forming an abscess to wall off the infection and facilitate bacterial clearance; the number of neutrophils that infiltrate an S. aureus skin infection often correlates with disease outcome. LysM-EGFP mice, which possess the enhanced green fluorescent protein (EGFP) inserted in the Lysozyme M (LysM) promoter region (expressed primarily by neutrophils), when used in conjunction with in vivo whole animal fluorescence imaging (FLI) provide a means of quantifying neutrophil emigration noninvasively and longitudinally into wounded skin. When combined with a bioluminescent S. aureus strain and sequential in vivo whole animal bioluminescent imaging (BLI), it is possible to longitudinally monitor both the neutrophil recruitment dynamics and in vivo bacterial burden at the site of infection in anesthetized mice from onset of infection to resolution or death. Mice are more resistant to a number of virulence factors produced by S. aureus that facilitate effective colonization and infection in humans. Immunodeficient mice provide a more sensitive animal model to examine persistent S. aureus infections and the ability of therapeutics to boost innate immune responses. Herein, we characterize responses in LysM-EGFP mice that have been bred to MyD88-deficient mice (LysM-EGFP×MyD88-/- mice) along with wild-type LysM-EGFP mice to investigate S. aureus skin wound infection. Multispectral simultaneous detection enabled study of neutrophil recruitment dynamics by using in vivo FLI, bacterial burden by using in vivo BLI, and wound healing longitudinally and noninvasively over time.


Subject(s)
Immunity, Innate/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Animals , Disease Models, Animal , Humans , Mice
14.
J Immunol ; 199(5): 1772-1782, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28733486

ABSTRACT

The immune response to Staphylococcus aureus infection in skin involves the recruitment of polymorphonuclear neutrophils (PMNs) from the bone marrow via the circulation and local granulopoiesis from hematopoietic stem and progenitor cells (HSPCs) that also traffic to infected skin wounds. We focus on regulation of PMN number and function and the role of pore-forming α-toxin (AT), a virulence factor that causes host cell lysis and elicits inflammasome-mediated IL-1ß secretion in wounds. Infection with wild-type S. aureus enriched in AT reduced PMN recruitment and resulted in sustained bacterial burden and delayed wound healing. In contrast, PMN recruitment to wounds infected with an isogenic AT-deficient S. aureus strain was unimpeded, exhibiting efficient bacterial clearance and hastened wound resolution. HSPCs recruited to infected wounds were unaffected by AT production and were activated to expand PMN numbers in proportion to S. aureus abundance in a manner regulated by TLR2 and IL-1R signaling. Immunodeficient MyD88-knockout mice infected with S. aureus experienced lethal sepsis that was reversed by PMN expansion mediated by injection of wild-type HSPCs directly into wounds. We conclude that AT-induced IL-1ß promotes local granulopoiesis and effective resolution of S. aureus-infected wounds, revealing a potential antibiotic-free strategy for tuning the innate immune response to treat methicillin-resistant S. aureus infection in immunodeficient patients.


Subject(s)
Bacterial Toxins/immunology , Granulocytes/immunology , Hematopoietic Stem Cells/physiology , Hemolysin Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Virulence Factors/immunology , Wound Infection/immunology , Animals , Bacterial Load , Bacterial Toxins/genetics , Cell Differentiation , Cell Proliferation , Granulocytes/microbiology , Hemolysin Proteins/genetics , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Myeloid Differentiation Factor 88/genetics , Receptors, Interleukin-1/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Virulence Factors/genetics
15.
Cell Death Differ ; 23(8): 1283-95, 2016 08.
Article in English | MEDLINE | ID: mdl-26891695

ABSTRACT

A-Raf belongs to the family of oncogenic Raf kinases that are involved in mitogenic signaling by activating the MEK-ERK pathway. Low kinase activity of A-Raf toward MEK suggested that A-Raf might have alternative functions. We recently identified A-Raf as a potent inhibitor of the proapoptotic mammalian sterile 20-like kinase (MST2) tumor suppressor pathway in several cancer entities including head and neck, colon, and breast. Independent of kinase activity, A-Raf binds to MST2 thereby efficiently inhibiting apoptosis. Here, we show that the interaction of A-Raf with the MST2 pathway is regulated by subcellular compartmentalization. Although in proliferating normal cells and tumor cells A-Raf localizes to the mitochondria, differentiated non-carcinogenic cells of head and neck epithelia, which express A-Raf at the plasma membrane. The constitutive or induced re-localization of A-Raf to the plasma membrane compromises its ability to efficiently sequester and inactivate MST2, thus rendering cells susceptible to apoptosis. Physiologically, A-Raf re-localizes to the plasma membrane upon epithelial differentiation in vivo. This re-distribution is regulated by the scaffold protein kinase suppressor of Ras 2 (KSR2). Downregulation of KSR2 during mammary epithelial cell differentiation or siRNA-mediated knockdown re-localizes A-Raf to the plasma membrane causing the release of MST2. By using the MCF7 cell differentiation system, we could demonstrate that overexpression of A-Raf in MCF7 cells, which induces differentiation. Our findings offer a new paradigm to understand how differential localization of Raf complexes affects diverse signaling functions in normal cells and carcinomas.


Subject(s)
Apoptosis , Cell Differentiation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins A-raf/metabolism , Caspase 8/metabolism , Cell Differentiation/drug effects , Cell Membrane/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Microscopy, Fluorescence , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuregulin-1/pharmacology , Proto-Oncogene Proteins A-raf/antagonists & inhibitors , Proto-Oncogene Proteins A-raf/genetics , RNA Interference , RNA, Small Interfering/metabolism , Serine-Threonine Kinase 3
16.
Eukaryot Cell ; 14(10): 983-97, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26209694

ABSTRACT

The global regulatory veA gene governs development and secondary metabolism in numerous fungal species, including Aspergillus flavus. This is especially relevant since A. flavus infects crops of agricultural importance worldwide, contaminating them with potent mycotoxins. The most well-known are aflatoxins, which are cytotoxic and carcinogenic polyketide compounds. The production of aflatoxins and the expression of genes implicated in the production of these mycotoxins are veA dependent. The genes responsible for the synthesis of aflatoxins are clustered, a signature common for genes involved in fungal secondary metabolism. Studies of the A. flavus genome revealed many gene clusters possibly connected to the synthesis of secondary metabolites. Many of these metabolites are still unknown, or the association between a known metabolite and a particular gene cluster has not yet been established. In the present transcriptome study, we show that veA is necessary for the expression of a large number of genes. Twenty-eight out of the predicted 56 secondary metabolite gene clusters include at least one gene that is differentially expressed depending on presence or absence of veA. One of the clusters under the influence of veA is cluster 39. The absence of veA results in a downregulation of the five genes found within this cluster. Interestingly, our results indicate that the cluster is expressed mainly in sclerotia. Chemical analysis of sclerotial extracts revealed that cluster 39 is responsible for the production of aflavarin.


Subject(s)
Aflatoxins/genetics , Aspergillus flavus/genetics , Fungal Proteins/genetics , Gene Expression Regulation, Fungal/genetics , Genes, Regulator/genetics , Multigene Family/genetics , Secondary Metabolism/genetics , Transcription Factors/genetics , Aflatoxins/biosynthesis , Aspergillus flavus/pathogenicity , Gene Expression Profiling , Transcriptome/genetics
17.
Br J Cancer ; 111(5): 955-64, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-24992580

ABSTRACT

BACKGROUND: Expression of epithelial cell adhesion molecule (EpCAM) is deregulated in epithelial malignancies. Beside its role in cell adhesion, EpCAM acts as signalling molecule with tumour-promoting functions. Thus, EpCAM is part of the molecular network of oncogenic receptors and considered an interesting therapeutic target. METHODS: Here, we thoroughly characterised EpCAM expression on mRNA and protein level in comprehensive tissue studies including non-cancerous prostate specimens, primary tumours of different grades and stages, metastatic lesions, and therapy-treated tumour specimens, as well as in prostate cancer cell lines. RESULTS: Epithelial cell adhesion molecule was overexpressed at mRNA and at protein level in prostate cancer tissues and cell lines. Altered EpCAM expression was an early event in prostate carcinogenesis with an upregulation in low-grade cancers and further induction in high-grade tumours and metastatic lesions. Interestingly, EpCAM was repressed upon induction of epithelial-to-mesenchymal transition (EMT) following chemotherapeutic treatment with docetaxel. Oppositely, re-induction of the epithelial phenotype through miRNAs miR-200c and miR-205, two inducers of mesenchymal-to-epithelial transition (MET), led to re-induction of EpCAM in chemoresistant cells. Furthermore, we prove that EpCAM cleavage, the first step of EpCAM signalling takes place in prostate cancer cells but in contrast to other cancer entities, EpCAM has no measurable impact on the proliferative behaviour of prostate cells, in vitro. CONCLUSIONS: In conclusion, our data confirm that EpCAM overexpression is an early event during prostate cancer progression. Epithelial cell adhesion molecule displays a dynamic, heterogeneous expression and associates with epithelial cells rather than mesenchymal, chemoresistant cells along with processes of EMT and MET.


Subject(s)
Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial Cell Adhesion Molecule , Humans , Male , MicroRNAs/drug effects , Prostatic Neoplasms/drug therapy , RNA, Messenger/genetics , Taxoids/pharmacology
18.
Oncogene ; 33(41): 4904-15, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-24141784

ABSTRACT

The role of the epithelial cell adhesion molecule EpCAM in cancer progression remains largely unclear. High expression of EpCAM in primary tumors is often associated with more aggressive phenotypes and EpCAM is the prime epithelial antigen in use to isolate circulating tumor cells (CTCs) and characterize disseminated tumor cells (DTCs). However, reduced expression of EpCAM was associated with epithelial-to-mesenchymal transition (EMT) and reports on a lack of EpCAM on CTCs emerged. These contradictory observations might reflect a context-dependent adaption of EpCAM expression during metastatic progression. To test this, EpCAM expression was monitored in esophageal cancer at different sites of early systemic disease. Although most of the primary esophageal tumors expressed high levels of EpCAM, the majority of DTCs in bone marrow lacked EpCAM. In vitro, downregulation of EpCAM expression at the plasma membrane was observed in migrating and invading cells, and was associated with a partial loss of the epithelial phenotype and with significantly decreased proliferation. Accordingly, induction of EMT through the action of TGFß resulted in substantial loss of EpCAM cell surface expression on esophageal cancer cells. Knock-down or natural loss of EpCAM recapitulated these effects as it reduced proliferation while enhancing migration and invasion of cancer cells. Importantly, expression of EpCAM on DTCs was significantly associated with the occurrence of lymph node metastases and with significantly decreased overall survival of esophageal cancer patients. We validated this observation by showing that high expression of EpCAM promoted tumor outgrowth after xenotransplantation of esophageal carcinoma cells. The present data disclose a dynamic expression of EpCAM throughout tumor progression, where EpCAM(high) phenotypes correlate with proliferative stages, whereas EpCAM(low/negative) phenotypes associated with migration, invasion and dissemination. Thus, differing expression levels of EpCAM must be taken into consideration for therapeutic approaches and during clinical retrieval of disseminated tumor cells.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Esophageal Neoplasms/pathology , Lymphatic Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Aged , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial Cell Adhesion Molecule , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Lymphatic Metastasis/genetics , Male , Mice , Mice, Inbred NOD , Middle Aged , Neoplastic Cells, Circulating/metabolism , Phenotype , Transforming Growth Factor beta/metabolism
19.
Oncogene ; 32(5): 641-50, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-22391566

ABSTRACT

The epithelial cell adhesion molecule (EpCAM) is an integral transmembrane protein that is frequently overexpressed in embryonic stem cells, tissue progenitors, carcinomas and cancer-initiating cells. In cancer cells, expression of EpCAM is associated with enhanced proliferation and upregulation of target genes including c-myc. However, the exact molecular mechanisms underlying the observed EpCAM-dependent cell proliferation remained unexplored. Here, we show that EpCAM directly affects cell cycle progression via its capacity to regulate the expression of cyclin D1 at the transcriptional level and depending on the direct interaction partner FHL2 (four-and-a-half LIM domains protein 2). As a result, downstream events such as phosphorylation of the retinoblastoma protein (Rb) and expression of cyclins E and A are similarly affected. In vivo, EpCAM expression strength and pattern are both positively correlated with the proliferation marker Ki67, high expression and nuclear localisation of cyclin D1, and Rb phosphorylation. Thus, EpCAM enhances cell cycle progression via the classical cyclin-regulated pathway.


Subject(s)
Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Cycle/genetics , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Cell Proliferation , Cyclin A/metabolism , Cyclin E/metabolism , Epithelial Cell Adhesion Molecule , Humans , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Phosphorylation , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism
20.
J Environ Qual ; 39(3): 1043-50, 2010.
Article in English | MEDLINE | ID: mdl-20400600

ABSTRACT

Acidity of water from abandoned underground mines decreases over time, and the rate of decrease can help formulate remediation approaches and treatment system designs. The objective of this study was to determine an overall acidity decay rate for above-drainage underground mines in northern West Virginia from a large data set of mines that were closed 50 to 70 yr ago. Water quality data were obtained from 30 Upper Freeport and 7 Pittsburgh coal seam mines in 1968, 1980, 2000, and 2006, and acidity decay curves were calculated. The mean decay constant, k, for Upper Freeport mines was 2.73 x 10(-2) yr(-1), with a 95% confidence interval of +/- 0.0052, whereas the k value for Pittsburgh mines was not significantly different at 4.26 x 10(-2) yr(-1) +/- 0.017. Acidity from the T&T mine, which was closed 12 yr ago, showed a k value of 11.25 x 10(-2) yr(-1). This higher decay rate was likely due to initial flushing of accumulated metal salts on reaction surfaces in the mine, rapid changes in mine hydrology after closure, and treatment. Although each site showed a specific decay rate (varying from 0.04 x 10(-2) yr(-1) to 13.1 x 10(-2) yr(-1)), the decay constants of 2.7 x 10(-2) yr(-1) to 4.3 x 10(-2) yr(-1) are useful for predicting water quality trends and overall improvements across a wide spectrum of abandoned underground mines. We found first-order decay models improve long-term prediction of acidity declines from above-drainage mines compared with linear or percent annual decrease models. These predictions can help to select water treatment plans and evaluate costs for these treatments over time.


Subject(s)
Acids , Mining , Water Movements , Environmental Monitoring , Seasons , Time Factors , Water Pollutants, Chemical , West Virginia
SELECTION OF CITATIONS
SEARCH DETAIL