57 results on '"Demetrius Moskophidis"'
Search Results
2. Dusp26 phosphatase regulates mitochondrial respiration and oxidative stress and protects neuronal cell death
- Author
-
Binnur Eroglu, Xiongjie Jin, Sadiki Deane, Bahadır Öztürk, Owen A. Ross, Demetrius Moskophidis, and Nahid F. Mivechi
- Subjects
Pharmacology ,Male ,Mice, Knockout ,Mice, 129 Strain ,Cell Death ,Dopaminergic Neurons ,Cell Respiration ,Neurodegenerative Diseases ,Parkinson Disease ,Cell Biology ,Mitochondria ,Cellular and Molecular Neuroscience ,Mice ,Oxidative Stress ,HEK293 Cells ,Cytoprotection ,Molecular Medicine ,Animals ,Dual-Specificity Phosphatases ,Humans ,Mitogen-Activated Protein Kinase Phosphatases ,Molecular Biology ,Cells, Cultured - Abstract
The dual specificity protein phosphatases (Dusps) control dephosphorylation of mitogen-activated protein kinases (MAPKs) as well as other substrates. Here, we report that Dusp26, which is highly expressed in neuroblastoma cells and primary neurons is targeted to the mitochondrial outer membrane via its NH
- Published
- 2021
3. The transcriptional regulator of the chaperone response HSF1 controls hepatic bioenergetics and protein homeostasis
- Author
-
Junfeng Pang, Xiongjie Jin, Demetrius Moskophidis, Nahid F. Mivechi, and Aijun Qiao
- Subjects
0301 basic medicine ,Bioenergetics ,Transcription, Genetic ,Nicotinamide phosphoribosyltransferase ,Regulator ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Adenosine Triphosphate ,Heat Shock Transcription Factors ,Animals ,Homeostasis ,HSF1 ,Research Articles ,Heat-Shock Proteins ,biology ,Cell Biology ,NAD ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,Proteostasis ,chemistry ,Biochemistry ,Gene Expression Regulation ,Liver ,Chaperone (protein) ,biology.protein ,NAD+ kinase ,Energy Metabolism ,Protein Processing, Post-Translational ,Heat-Shock Response ,Deacetylase activity ,Molecular Chaperones ,Transcription Factors - Abstract
How cells sense energetic demands and regulate their bioenergetic networks to balance anabolism and catabolism is unclear. Qiao et al show that HSF1, a regulator of the chaperone response, has a central role in systemic energy sensing and is required for metabolic adaptation to nutrient availability., Metabolic energy reprogramming facilitates adaptations to a variety of stress conditions and cellular dysfunction, but how the energetic demands are monitored and met in response to physiological stimuli remains elusive. Our data support a model demonstrating that heat shock factor 1 (HSF1), a master transcriptional regulator of the chaperone response, has been coopted from its role as a critical protein quality-control regulator to having a central role in systemic energy sensing and for metabolic adaptation to nutrient availability. We found that in the absence of HSF1, levels of NAD+ and ATP are not efficiently sustained in hepatic cells, largely because of transcriptional repression of nicotinamide phosphoribosyltransferase in the NAD+ salvage pathway. Mechanistically, the defect in NAD+ and ATP synthesis linked to a loss of NAD+-dependent deacetylase activity, increased protein acetylation, and impaired mitochondrial integrity. Remarkably, the drop in ATP level caused by HSF1 loss invoked an adaptive response featuring the inhibition of energetically demanding processes, including gluconeogenesis, translation, and lipid synthesis. Our work identifies HSF1 as a central regulator of cellular bioenergetics and protein homeostasis that benefits malignant cell progression and exacerbates development of metabolic diseases.
- Published
- 2017
4. HSF1-mediated Control of Cellular Energy Metabolism and mTORC1 Activation Drive Acute T Cell Lymphoblastic Leukemia Progression
- Author
-
Caixia Xi, Nahid F. Mivechi, Demetrius Moskophidis, Junfeng Pang, Xiongjie Jin, and Binnur Eroglu
- Subjects
0301 basic medicine ,Male ,Cancer Research ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Heat Shock Transcription Factors ,Cell Line, Tumor ,Medicine ,PTEN ,Animals ,Humans ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Acute leukemia ,biology ,business.industry ,fungi ,medicine.disease ,Leukemia ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Disease Progression ,Female ,Signal transduction ,business ,Energy Metabolism ,Signal Transduction - Abstract
Deregulated oncogenic signaling linked to PI3K/AKT and mTORC1 pathway activation is a hallmark of human T-cell acute leukemia (T-ALL) pathogenesis and contributes to leukemic cell resistance and adverse prognosis. Notably, although the multiagent chemotherapy of leukemia leads to a high rate of complete remission, options for salvage therapy for relapsed/refractory disease are limited due to the serious side effects of augmenting cytotoxic chemotherapy. We report that ablation of HSF1, a key transcriptional regulator of the chaperone response and cellular bioenergetics, from mouse T-ALL tumors driven by PTEN loss or human T-ALL cell lines, has significant therapeutic effects in reducing tumor burden and sensitizing malignant cell death. From a mechanistic perspective, the enhanced sensitivity of T-ALLs to HSF1 depletion resides in the reduced MAPK–ERK signaling and metabolic and ATP-producing capacity of malignant cells lacking HSF1 activity. Impaired mitochondrial ATP production and decreased intracellular amino acid content in HSF1-deficient T-ALL cells trigger an energy-saving adaptive response featured by attenuation of the mTORC1 activity, which is coregulated by ATP, and its downstream target proteins (p70S6K and 4E-BP). This leads to protein translation attenuation that diminishes oncogenic signals and malignant cell growth. Collectively, these metabolic alterations in the absence of HSF1 activity reveal cancer cell liabilities and have a profound negative impact on T-ALL progression. Implications: Targeting HSF1 and HSF1-dependent cancer-specific anabolic and protein homeostasis programs has a significant therapeutic potential for T-ALL and may prevent progression of relapsed/refractory disease.
- Published
- 2019
5. The Molecular Chaperone Heat Shock Protein 70 Controls Liver Cancer Initiation and Progression by Regulating Adaptive DNA Damage and Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Signaling Pathways
- Author
-
Junfeng Pang, Yan Wang, Nahid F. Mivechi, Demetrius Moskophidis, Xiongjie Jin, and Wonkyoung Cho
- Subjects
Male ,MAPK/ERK pathway ,Carcinoma, Hepatocellular ,MAP Kinase Signaling System ,Tumor initiation ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Diethylnitrosamine ,HSP70 Heat-Shock Proteins ,Protein kinase A ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Liver cell ,Liver Neoplasms ,Cell Biology ,Cell cycle ,Cell Transformation, Neoplastic ,Tumor progression ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Mitogen-activated protein kinase ,Disease Progression ,Cancer research ,biology.protein ,Tumor Suppressor Protein p53 ,Signal transduction ,DNA Damage ,Research Article - Abstract
Delineating the mechanisms that drive hepatic injury and hepatocellular carcinoma (HCC) progression is critical for development of novel treatments for recurrent and advanced HCC but also for the development of diagnostic and preventive strategies. Heat shock protein 70 (HSP70) acts in concert with several cochaperones and nucleotide exchange factors and plays an essential role in protein quality control that increases survival by protecting cells against environmental stressors. Specifically, the HSP70-mediated response has been implicated in the pathogenesis of cancer, but the specific mechanisms by which HSP70 may support malignant cell transformation remains to be fully elucidated. Here, we show that genetic ablation of HSP70 markedly impairs HCC initiation and progression by distinct but overlapping pathways. This includes the potentiation of the carcinogen-induced DNA damage response, at the tumor initiation stage, to increase the p53-dependent surveillance response leading to the cell cycle exit or death of genomically damaged differentiated pericentral hepatocytes, and this may also prevent their conversion into more proliferating HCC progenitor cells. Subsequently, activation of a mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) negative feedback pathway diminishes oncogenic signals, thereby attenuating premalignant cell transformation and tumor progression. Modulation of HSP70 function may be a strategy for interfering with oncogenic signals driving liver cell transformation and tumor progression, thus providing an opportunity for human cancer control.
- Published
- 2019
6. Role of heat shock factor 1 (HSF1) on activation-induced metabolic reprogramming of T cells
- Author
-
Bhaumik D Pandya, Demetrius Moskophidis, and Nahid F Mivechi
- Subjects
Immunology ,Immunology and Allergy - Abstract
Heat Shock Factor 1 (HSF1) is transcriptionally activated in response to a variety of environmental stressors. Data from our laboratory indicates that genetic inactivation of HSF1 significantly delays tumorigenesis in various tumor models. Increasing evidence supports the paradigm that modulating the metabolic bioenergetics of T cells i.e., inhibiting glycolytic flux or enforcing fatty acid oxidation, enhances the formation of more efficient anti tumor CD8+ T-cells. Since, HSF1 is involved in cellular metabolism, in this study, we explored the strategies to manipulate (or reprogram) the metabolic pathways used by the immune system to improve anti-tumor immunity. We observed that deletion of HSF1 profoundly affects the metabolic reprogramming of naive CD8+ T cells upon anti-CD3/CD28 stimulation in vitro. Furthermore, HSF1-deficient T cells show reduced oxygen consumption rate and extracellular acidification rate upon activation. These effects correspond to delayed T-cell receptor signaling and slower activation of naive T cells preventing them from moving towards early exhaustion stage resulting in improved longevity. In addition, we detected delayed initiation of colon cancer MC38 tumors that was implanted into a T-cell specific HSF1 deficient mice compared to wild-type mice. Finally, we confirmed that the delay in tumorigenesis is mediated through CD8 response since treatment of above tumor bearing mice with anti-CD8 antibody reversed the suppressive effects of HSF1 deficient CD8 T cells on tumor growth. Taken together, HSF1 can be a potential therapeutic target to overcome the limitations with existing cancer therapies and further studies will be directed towards exploring impact of HSF1 on checkpoint blockade.
- Published
- 2020
7. Modulation of Heat Shock Factor 1 Activity through Silencing of Ser303/Ser307 Phosphorylation Supports a Metabolic Program Leading to Age-Related Obesity and Insulin Resistance
- Author
-
Xiongjie Jin, Aijun Qiao, Demetrius Moskophidis, and Nahid F. Mivechi
- Subjects
0301 basic medicine ,Male ,Aging ,medicine.medical_treatment ,Mice, Transgenic ,03 medical and health sciences ,Mice ,Insulin resistance ,Heat Shock Transcription Factors ,medicine ,Transcriptional regulation ,Serine ,Gene silencing ,Animals ,Humans ,Gene Knock-In Techniques ,Obesity ,Phosphorylation ,HSF1 ,Molecular Biology ,Cells, Cultured ,biology ,Protein Stability ,Growth factor ,fungi ,Biological activity ,Cell Biology ,medicine.disease ,Recombinant Proteins ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Amino Acid Substitution ,Chaperone (protein) ,biology.protein ,Female ,Insulin Resistance ,Heat-Shock Response ,Research Article - Abstract
Activation of the adaptive response to cellular stress orchestrated by heat shock factor 1 (HSF1), which is an evolutionarily conserved transcriptional regulator of chaperone response and cellular bioenergetics in diverse model systems, is a central feature of organismal defense from environmental and cellular stress. HSF1 activity, induced by proteostatic, metabolic, and growth factor signals, is regulated by posttranscriptional modifications, yet the mechanisms that regulate HSF1 and particularly the functional significance of these modifications in modulating its biological activity in vivo remain unknown. HSF1 phosphorylation at both Ser303 (S303) and Ser307 (S307) has been shown to repress HSF1 transcriptional activity under normal physiological growth conditions. To determine the biological relevance of these HSF1 phosphorylation events, we generated a knock-in mouse model in which S303 and S307 were replaced with alanine (HSF1(303A/307A)). Our results confirmed that loss of phosphorylation in HSF1(303A/307A) cells and tissues increases protein stability but also markedly sensitizes HSF1 activation under normal and heat- or nutrient-induced stress conditions. Interestingly, the enhanced HSF1 activation in HSF1(303A/307A) mice activates a supportive metabolic program that aggravates the development of age-dependent obesity, fatty liver diseases, and insulin resistance. Thus, these findings highlight the importance of a posttranslational mechanism (through phosphorylation at S303 and S307 sites) of regulation of the HSF1-mediated transcriptional program that moderates the severity of nutrient-induced metabolic diseases.
- Published
- 2018
8. Heat Shock Factor Hsf1 Cooperates with ErbB2 (Her2/Neu) Protein to Promote Mammary Tumorigenesis and Metastasis
- Author
-
Demetrius Moskophidis, Yanzhong Hu, Nahid F. Mivechi, Phillip Buckhaults, and Caixia Xi
- Subjects
MAP Kinase Signaling System ,Receptor, ErbB-2 ,Mammary Neoplasms, Animal ,Vimentin ,Biochemistry ,HER2/neu ,Metastasis ,Mice ,Heat Shock Transcription Factors ,Transforming Growth Factor beta ,medicine ,Animals ,HSP90 Heat-Shock Proteins ,Neoplasm Metastasis ,HSF1 ,Molecular Biology ,Mice, Knockout ,Mitogen-Activated Protein Kinase 3 ,biology ,fungi ,Cell Biology ,Transforming growth factor beta ,Cadherins ,medicine.disease ,Hsp90 ,Molecular biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Proto-Oncogene Proteins c-raf ,Heat shock factor ,Cell Transformation, Neoplastic ,biology.protein ,Cancer research ,Female ,Signal transduction ,Transcription Factors - Abstract
ErbB2/Neu oncogene is overexpressed in 25% of invasive/metastatic breast cancers. We have found that deletion of heat shock factor Hsf1 in mice overexpressing ErbB2/Neu significantly reduces mammary tumorigenesis and metastasis. Hsf1(+/-)ErbB2/Neu(+) tumors exhibit reduced cellular proliferative and invasive properties associated with reduced activated ERK1/2 and reduced epithelial-mesenchymal transition (EMT). Hsf1(+/+)Neu(+) mammary epithelial cells exposed to TGFβ show high levels of ERK1/2 activity and EMT; this is associated with reduced expression of E-cadherin and increased expression of Slug and vimentin, a mesenchymal marker. In contrast, Hsf1(-/-)Neu(+) or Hsf1(+/+)Neu(+) cells do not exhibit activated ERK1/2 and show reduced EMT in the presence of TGFβ. The ineffective activation of the RAS/RAF/MEK/ERK1/2 signaling pathway in cells with reduced levels of HSF1 is due to the low levels of HSP90 in complex with RAF1 that are required for RAF1 stability and maturation. These results indicate a powerful inhibitory effect conferred by HSF1 downstream target genes in the inhibition of ErbB2-induced breast cancers in the absence of the Hsf1 gene.
- Published
- 2012
9. Heat Shock Transcription Factor 1 Is a Key Determinant of HCC Development by Regulating Hepatic Steatosis and Metabolic Syndrome
- Author
-
Xiongjie Jin, Demetrius Moskophidis, and Nahid F. Mivechi
- Subjects
medicine.medical_specialty ,Carcinoma, Hepatocellular ,Physiology ,AMP-Activated Protein Kinases ,Article ,Mice ,Insulin resistance ,Heat Shock Transcription Factors ,AMP-activated protein kinase ,Internal medicine ,Lipid biosynthesis ,medicine ,Animals ,HSF1 ,Molecular Biology ,Metabolic Syndrome ,Mice, Knockout ,biology ,Liver Neoplasms ,fungi ,Fatty liver ,Lipid metabolism ,Cell Biology ,Lipid Metabolism ,medicine.disease ,Receptor, Insulin ,DNA-Binding Proteins ,Fatty Liver ,Heat shock factor ,Endocrinology ,biology.protein ,Steatosis ,Signal Transduction ,Transcription Factors - Abstract
SummaryHepatocellular carcinoma (HCC) occurrence and progression are linked tightly to progressive hepatic metabolic syndrome associated with insulin resistance, hepatic steatosis, and chronic inflammation. Heat shock transcription factor 1 (HSF1), a major transactivator of stress proteins, increases survival by protecting cells against environmental stressors. It has been implicated in the pathogenesis of cancer, but specific mechanisms by which HSF1 supports cancer development remain elusive. We propose a pathogenic mechanism whereby HSF1 activation promotes growth of premalignant cells and HCC development by stimulating lipid biosynthesis and perpetuating chronic hepatic metabolic disease induced by carcinogens. Our work shows that inactivation of HSF1 impairs cancer progression, mitigating adverse effects of carcinogens on hepatic metabolism by enhancing insulin sensitivity and sensitizing activation of AMP-activated protein kinase (AMPK), an important regulator of energy homeostasis and inhibitor of lipid synthesis. HSF1 is a potential target for the control of hepatic steatosis, hepatic insulin resistance, and HCC development.
- Published
- 2011
- Full Text
- View/download PDF
10. Loss of Hsp110 Leads to Age-Dependent Tau Hyperphosphorylation and Early Accumulation of Insoluble Amyloid β
- Author
-
Binnur Eroglu, Nahid F. Mivechi, and Demetrius Moskophidis
- Subjects
Male ,Aging ,Amyloid ,Green Fluorescent Proteins ,Immunoblotting ,Tau protein ,Mice, Transgenic ,tau Proteins ,Motor Activity ,Biology ,Amyloid beta-Protein Precursor ,Mice ,GSK-3 ,Heat shock protein ,mental disorders ,medicine ,Amyloid precursor protein ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Protein Phosphatase 2 ,Senile plaques ,HSP110 Heat-Shock Proteins ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,Neurons ,Amyloid beta-Peptides ,Microscopy, Confocal ,Neurodegeneration ,Brain ,Articles ,Cell Biology ,medicine.disease ,Immunohistochemistry ,Solubility ,Biochemistry ,biology.protein ,PIN1 ,Female ,Amyloid Precursor Protein Secretases ,Protein Binding - Abstract
Diseases like Alzheimer's disease (AD) and other tauopathies are defined by the expression of neurofibrillary tangles (NFTs) deposited mainly in neurons. The NFTs are aggregates of the hyperphosphorylated tau (p-tau) (3, 74). Normal tau increases microtubule stability, but tau can be hyperphosphorylated under disease conditions and released from microtubules (3, 5, 6). The molecular mechanisms involved in the formation of NFTs are not completely understood. However, accumulation of abnormal p-tau and NFTs causes neurodegeneration (3). A number of protein kinases, including glycogen synthase kinase 3 (GSK3) and cyclin-dependent protein kinase 5 (CDK5), have been shown to phosphorylate tau at Thr231 and Ser262 as well as several other sites that flank the microtubule binding repeat, leading to tangles of paired helical filaments (PHFs) similar to those observed in the brains of patients with AD (54, 72). Evidence shows that GSK3 physically interacts with tau and is thought to be the main contributor to the formation of NFTs and amyloid β (Aβ) plaques in AD patients (18, 53, 54). Phosphorylation of GSK3a/b at S9/S21 which is inhibitory to its activity during insulin signaling, leads to phosphorylation of tau in neurons (80). GSK3a/b phospho-S9/S21, p-tau, and 14-3-3zeta have been isolated in a 500-kDa complex, and the interaction has been shown to result in tau phosphorylation by GSK3 (1, 80). Although not well characterized, p-tau has been shown to be dephosphorylated by the B family regulatory subunit of the heterotrimeric PP2A holoenzyme (76). There are two protein phosphatase 2A (PP2A) binding sites on microtubule tau binding repeats, perhaps allowing tau to be more efficiently dephosphorylated by PP2A catalytic subunit (76). Both GSK3 and CDK5 are also known to be involved in the phosphorylation of amyloid precursor protein (APP) at Thr668 and APP processing and Aβ production (53, 58). Studies suggest that amyloid peptide can activate GSK3 signaling, and the increase in GSK3 activity can then contribute to abnormal APP processing. Indeed, reduction in GSK3 activity reduces amyloid peptide production in murine AD models (18, 53, 57, 71). Reduction in PP2A activity leads to altered APP regulation as well (26, 43). Additional molecules that affect tau hyperphosphorylation and APP processing are the peptidyl prolyl isomerases (9, 36, 51). Deletion of Pin1 isomerase in vivo leads to p-tau and neurodegeneration (42). Crossing Pin1-deficient mice with transgenic mice expressing mutant APP (APPβsw) leads to abnormal APP processing and accumulation of toxic amyloid β42 (Aβ42) species. Pin1, therefore, is implicated in isomerization of tau, perhaps facilitating its dephosphorylation (42). The presence of Pin1 has been implicated in promoting nonamyloidogenic processing of APP and reduction in toxic Aβ42 production (51). Hsp70/Hsc70 has been shown to preferentially bind to a hyperphosphorylated form of tau in the diseased human brain (49). Cross talk between the ubiquitin proteasome system (UPS) and molecular chaperones might also be critical in regulating the deposition and toxicity of tau (8, 16). These results suggest that the activity of Hsp70 and Hsp90 preserve the native structure and function of tau protein. Hsp70 and the C-terminal Hsp70-interacting protein (Chip) have been shown to regulate tau ubiquitination and degradation (11, 12, 21, 52, 65). Interestingly, Chip and βAPP interact, and Chip and Hsp70/90 expression have been shown to lower the cellular levels of Aβ and reduce Aβ toxicity in vitro (39). Misfolded proteins are either degraded through the UPS or are folded, at least in part, by the Hsps (4, 7). Eukaryotic cells possess a class of heat shock proteins (Hsps) related to the Hsp70 family. This Hsp100 family of proteins contains Hspa41 (Apg1 or OSP94), Hsp94 (Apg2), and Hsp110 (2, 17, 28, 61, 70, 77, 78). They were initially considered to be “holdases” that keep denatured proteins in solution, and no client proteins have been described for them (14, 15, 56, 62). Hsp110 interacts with Hsp70 and increases its ATPase activity (15, 56, 62). The main function of Hsp110 appears to be a nucleotide exchange factor (NEF) for Hsp70 (14, 64). In general, Hsp110 is known to induce suppression of aggregation and protein refolding, and it protects proteins from the damaging effects of various stresses; however, its physiological function in mammalian cells remains unknown (15, 60). In these studies, we examined the role of Hsp110 in central nervous system (CNS) homeostasis in vivo. We have found that hsp110−/− mice exhibit an age-dependent accumulation of p-tau that is associated with pathological features, such as the appearance of NFTs and neurodegeneration. We also show that lack of Hsp110 leads to accelerated pathology as evidenced by the early appearance of senile plaques containing Aβ42 (a major toxic species [46]) in an AD transgenic mouse model. At the biochemical level, we show that Hsp110 interacts with tau, a number of Hsps, GSK3, Pin1, and PP2A. Furthermore, tau immunocomplexes pulled down from hsp110−/− brain extracts possess elevated levels of PP2A, but the pulled-down PP2A has significantly lower activity than the PP2A from wild-type mice. Our studies therefore suggest a critical role for Hsp110 in maintaining the proper folding environment that is required for phosphorylation and dephosphorylation of tau and APP processing in vivo.
- Published
- 2010
11. Heat shock factor 1 deficiency via its downstream target gene αB-crystallin (Hspb5) impairs p53 degradation
- Author
-
Xiongjie Jin, Yanzhong Hu, Demetrius Moskophidis, Nahid F. Mivechi, and Andrew C. Phillips
- Subjects
Immunoprecipitation ,Protein degradation ,Transfection ,Biochemistry ,F-box protein ,Article ,Mice ,Heat Shock Transcription Factors ,Heat shock protein ,Animals ,HSF1 ,Molecular Biology ,Mice, Knockout ,biology ,F-Box Proteins ,fungi ,alpha-Crystallin B Chain ,Cell Biology ,Molecular biology ,Ubiquitin ligase ,DNA-Binding Proteins ,Heat shock factor ,biology.protein ,Tumor Suppressor Protein p53 ,Transcription Factors - Abstract
Heat shock factor Hsf1 regulates the stress-inducibility of heat shock proteins (Hsps) or molecular chaperones. One of the functions attributed to Hsps is their participation in folding and degradation of proteins. We recently showed that hsf1−/− cells accumulate ubiquitinated proteins. However, a direct role for Hsf1 in stability of specific proteins such as p53 has not been elucidated. We present evidence that cells deficient in hsf1 accumulate wild-type p53 protein. We further show that hsf1−/− cells express lower levels of αB-crystallin and cells deficient in αB-crystallin also accumulate p53 protein. Reports indicate that αB-crystallin binds to Fbx4 ubiquitin ligase, and they target cyclin D1 for degradation through a pathway involving the SCF (Skp1-Cul1-F-box) complex. Towards determining a mechanism for p53 degradation involving αB-crystallin and Hsf1, we have found that ectopic expression of Fbx4 in wild-type mouse embryo fibroblasts (MEFs) expressing mutant p53 (p53R175H) leads to increase in its degradation, while MEFs deficient in hsf1 or αBcry are defective in degradation of this p53 protein. In addition, immunoprecipitated p53R175H from wild-type MEFs is able to pull-down both αB-crystallin and Fbx4. Finally, immunoprecipitated wild-type p53 from doxorubicin treated U2OS cells can pull-down endogenous αB-crystallin and Fbx4. These results indicate that hsf1- and αBcry-deficient cells accumulate p53 due to reduced levels of αB-crystallin in these cells. Elevated levels of p53 in hsf1- and αBcry-deficient cells lead to their increased sensitivity to DNA damaging agents. These data reveal a novel mechanism for protein degradation through Hsf1 and αB-crystallin. J. Cell. Biochem. 107: 504–515, 2009. © 2009 Wiley-Liss, Inc.
- Published
- 2009
12. Control of Virus-Specific CD8+T-Cell Exhaustion and Immune-Mediated Pathology by E3 Ubiquitin Ligase Cbl-b during Chronic Viral Infection
- Author
-
Menghua Zhang, Lei Huang, Rong Ou, and Demetrius Moskophidis
- Subjects
Adoptive cell transfer ,Immunology ,Population ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocytic choriomeningitis ,Microbiology ,Lymphocyte Depletion ,Virus ,Mice ,Immune system ,Virology ,medicine ,Animals ,Arenaviridae Infections ,Lymphocytic choriomeningitis virus ,Cytotoxic T cell ,Aspartate Aminotransferases ,Proto-Oncogene Proteins c-cbl ,education ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Mice, Knockout ,education.field_of_study ,Alanine Transaminase ,medicine.disease ,Adoptive Transfer ,Survival Analysis ,Ubiquitin ligase ,Mice, Inbred C57BL ,Liver ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,CD8 - Abstract
A characteristic feature in the immune response to many persistent viral infections is the dysfunction or deletion of antigen-specific T cells (exhaustion). This down-regulation of virus-specific T-cell response represents a critical control mechanism that exists within T-cell activation pathways to prevent lethal disease by inappropriate responses against disseminating virus infections. However, the molecular mechanisms by which the immune system determines whether to mount a full response to such infections remain largely unexplored. Here, we have established that in the murine lymphocytic choriomeningitis virus (LCMV) model, induction of the T-cell receptor signaling inhibitor molecule E3 ligase Cbl-b is critically involved in this decision. In particular, our data revealed that Cbl-b controls the program responsible for T-cell tolerance (exhaustion) induction during a chronic viral infection. Thus, Cbl-b−/−mice infected with a low dose of LCMV Docile mount a strong CD8+T-cell response that rapidly clears the infection, and the animals remain healthy; in contrast, down-regulation of the epitope-specific CD8+T-cell population in persistently infected Cbl-b−/−mice, compared to that in chronically infected B6 mice, was significantly delayed, and this was associated with increased morbidity and eventual death in nearly 20% of the animals. Interestingly, infection of Cbl-b−/−mice with a moderate virus dose resulted in rapid death with 100% mortality by 7 to 8 days after infection, caused by a dysregulated antiviral T-cell response, whereas the infected B6 mice survived and remained healthy. In conclusion, our results suggest that Cbl-b is critically involved in T-cell exhaustion and prevention of lethal disease.
- Published
- 2008
13. Regulation of Immune Response and Inflammatory Reactions against Viral Infection by VCAM-1
- Author
-
Menghua Zhang, Demetrius Moskophidis, Pandelakis A. Koni, Richard A. Flavell, Rong Ou, and Lei Huang
- Subjects
Immunology ,Vascular Cell Adhesion Molecule-1 ,Mice, Transgenic ,Inflammation ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Biology ,Antibodies, Viral ,Virus Replication ,Lymphocytic choriomeningitis ,Microbiology ,Mice ,chemistry.chemical_compound ,Immune system ,Virology ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,VCAM-1 ,Cell adhesion molecule ,medicine.disease ,Mice, Inbred C57BL ,Viral replication ,chemistry ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,medicine.symptom ,Antibody ,CD8 - Abstract
The migration of activated antigen-specific immune cells to the target tissues of virus replication is controlled by the expression of adhesion molecules on the vascular endothelium that bind to ligands on circulating lymphocytes. Here, we demonstrate that the adhesion pathway mediated by vascular cell adhesion molecule 1 (VCAM-1) plays a role in regulating T-cell-mediated inflammation and pathology in nonlymphoid tissues, including the central nervous system (CNS) during viral infection. The ablation of VCAM-1 expression from endothelial and hematopoietic cells using aloxP-Crerecombination strategy had no major effect on the induction or overall tissue distribution of antigen-specific T cells during a systemic infection with lymphocytic choriomeningitis virus (LCMV), except in the case of lung tissue. However, enhanced resistance to lethal LCM and the significantly reduced magnitude and duration of footpad swelling observed in VCAM-1 mutant mice compared to B6 controls suggest a significant role for VCAM-1 in promoting successful local inflammatory reactions associated with efficient viral clearance and even life-threatening immunopathology under particular infection conditions. Interestingly, analysis of the infiltrating populations in the brains of intracerebrally infected mice revealed that VCAM-1 deletion significantly delayed migration into the CNS of antigen-presenting cells (macrophages and dendritic cells), which are critical for optimal stimulation of migrating virus-specific CD8+T cells initiating a pathological cascade. We propose that the impaired migration of these accessory cells in the brain may explain the improved clinical outcome of infection in VCAM-1 mutant mice. Thus, these results underscore the potential role of VCAM-1 in regulating the immune response and inflammatory reactions against viral infections.
- Published
- 2008
14. Insights into function and regulation of small heat shock protein 25 (HSPB1) in a mouse model with targeted gene disruption
- Author
-
Demetrius Moskophidis, Lei Huang, Nahid F. Mivechi, Shane C. Masters, and Jin Na Min
- Subjects
Male ,Cell physiology ,animal structures ,Fever ,Blotting, Western ,Apoptosis ,Stratified squamous epithelium ,Biology ,Mice ,Endocrinology ,Bone Marrow ,Radiation, Ionizing ,Heat shock protein ,Genetics ,medicine ,Animals ,Myocyte ,Muscle, Skeletal ,Gene ,Crosses, Genetic ,Heat-Shock Proteins ,Etoposide ,Mice, Inbred BALB C ,Integrases ,Gene Expression Regulation, Developmental ,Cell Biology ,Embryo, Mammalian ,beta-Galactosidase ,Antineoplastic Agents, Phytogenic ,Molecular biology ,Neoplasm Proteins ,Cell biology ,Hsp70 ,Mice, Inbred C57BL ,Heat shock factor ,Blotting, Southern ,Blastocyst ,medicine.anatomical_structure ,Lac Operon ,Gene Targeting ,Knockout mouse ,Female ,Molecular Chaperones - Abstract
The mammalian small heat shock protein (sHSPs) family is comprised of 10 members and includes HSPB1, which is proposed to play an essential role in cellular physiology, acting as a molecular chaperone to regulate diverse cellular processes. Whilst differential roles for sHSPs are suggested for specific tissues, the relative contribution of individual sHSP family members in cellular and organ physiology remains unclear. To address the function of HSPB1 in vivo and determine its tissue-specific expression during development and in the adult, we generated knock-in mice where the coding sequence of hspb1 is replaced by a lacZ reporter gene. Hspb1 expression marks myogenic differentiation with specific expression first confined to developing cardiac muscles and the vascular system, and later in skeletal muscles with specific expression at advanced stages of myoblast differentiation. In the adult, hspb1 expression was observed in other tissues, such as stratified squamous epithelium of skin, oronasal cavity, tongue, esophagus, and uterine cervix but its expression was most prominent in the musculature. Interestingly, in cardiac muscle hsbp1 expression was down-regulated during the neonatal period and maintained to a relatively low steady-level throughout adulthood. Despite this widespread expression, hspb1-/- mice were viable and fertile with no apparent morphological abnormalities in tissues under physiological conditions. However, at the cellular level and under stress conditions (heat challenge), HSPB1 act synergistically with the stress-induced HSPA1 (HSP70) in thermotolerance development, protecting cells from apoptosis. Our data thus indicate a nonessential role for HSPB1 in embryonic development and for maintenance of tissues under physiological conditions, but also shows that it plays an important role by acting synergistically with other HSPs during stress conditions to exert cytoprotection and anti-apoptotic effects.
- Published
- 2007
15. Perforin and Fas Cytolytic Pathways Coordinately Shape the Selection and Diversity of CD8+-T-Cell Escape Variants of Influenza Virus
- Author
-
Graeme E. Price, Menghua Zhang, Demetrius Moskophidis, Rong Ou, and Lei Huang
- Subjects
Pore Forming Cytotoxic Proteins ,Fas Ligand Protein ,viruses ,Immunology ,Orthomyxoviridae ,Mice, Transgenic ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Microbiology ,Antigenic drift ,Virus ,Mice ,Virology ,Antigenic variation ,Animals ,Cytotoxic T cell ,fas Receptor ,DNA Primers ,Homeodomain Proteins ,Mice, Knockout ,Membrane Glycoproteins ,Base Sequence ,biology ,Perforin ,Genetic Variation ,hemic and immune systems ,biology.organism_classification ,CTL ,Granzyme ,Insect Science ,biology.protein ,Pathogenesis and Immunity - Abstract
Antigenic variation is a viral strategy exploited to promote survival in the face of the host immune response and represents a major challenge for efficient vaccine development. Influenza viruses are pathogens with high transmissibility and mutation rates, enabling viral escape from immunity induced by prior infection or vaccination. Intense selection from neutralizing antibody drives antigenic changes in the surface glycoproteins, resulting in emergence of new strains able to reinfect hosts immune to previously circulating viruses. CD8+cytotoxic T cells (CTLs) also provide protective immunity from influenza virus infection and may contribute to the antigenic evolution of influenza viruses. Utilizing mice transgenic for an influenza virus NP366-374 peptide-specific T-cell receptor, we demonstrated that the respiratory tract is a suitable site for generation of escape variants of influenza virus selected by CTL in vivo. In this report the contributions of the perforin and Fas pathways utilized by influenza virus-specific CTLs in viral clearance and selection of CTL escape variants have been evaluated. While transgenic CTLs deficient in either perforin- or Fas-mediated pathways are efficient in initial pulmonary viral control, variant virus emergence was observed in all the mice studied, although the spectrum of viral CTL escape variants selected varied profoundly. Thus, a less-restricted repertoire of escape variants was observed in mice with an intact perforin cytotoxic pathway compared with a limited variant diversity in perforin pathway-deficient mice, although maximal variant diversity was observed in mice having both Fas and perforin pathways intact. We conclude that selection of viral CTL escape variants reflects coordinate action between the tightly controlled perforin/granzyme pathway and the more promiscuous Fas/FasL pathway.
- Published
- 2005
16. Targeted disruption of the heat shock transcription factor (hsf)-2 gene results in increased embryonic lethality, neuronal defects, and reduced spermatogenesis
- Author
-
Demetrius Moskophidis, Nahid F. Mivechi, Guanghu Wang, and Jing Zhang
- Subjects
Central Nervous System ,Male ,Molecular Probe Techniques ,Biology ,Mice ,Transactivation ,Endocrinology ,Heat shock protein ,Testis ,Genetics ,Animals ,Fetal Viability ,Spermatogenesis ,HSF1 ,Heat-Shock Proteins ,DNA Primers ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Embryogenesis ,Chromosome Mapping ,Gene targeting ,Embryo ,Cell Biology ,Flow Cytometry ,Cell biology ,Heat shock factor ,Gene Expression Regulation ,Immunology ,Knockout mouse ,Female ,Transcription Factors - Abstract
Summary: Heat shock transcription factors (Hsfs) are major transactivators of heat shock protein (Hsp) genes in the response to stress stimuli, but are also thought to be involved in embryonic development and spermatogenesis. Among the three known mammalian Hsfs, Hsf1 is recognized as the most effective transactivator of Hsps in response to thermal challenge, but the role of Hsf2 in regulation of genes under normal or increased stress conditions in vivo remains elusive. To study its physiological function in vivo, we generated mice deficient in hsf2 by gene targeting. We report here that hsf2−/− mice exhibit multiple phenotypes, including an increased prenatal lethality occurring between mid-gestation to birth, with fetal death probably due to central nervous system defects including collapse of the lateral ventricles and ventricular hemorrhages. Approximately 30% of hsf2−/− animals surviving to adulthood exhibited brain abnormalities characterized by marked dilation of the third and lateral ventricles. In addition, disruption of hsf2 resulted in reduced female fertility; however, despite ubiquitous expression in the testes and markedly reduced testis size and sperm count, only a small reduction in fertility was apparent in hsf2−/− male mice. Immunoblotting and gene expression microarray analysis of hsf2−/− embryos did not reveal reduced Hsp expression levels, indicating that the defects observed in hsf2−/− embryos may not result from disruption of Hsp expression. These findings suggest that hsf2 has a major function in controlling expression of genes important for embryonic development and maintenance of sperm production. genesis 36:48–61, 2003. © 2003 Wiley-Liss, Inc.
- Published
- 2003
17. WITHDRAWN: Abrogation of heat shock factor 1 (Hsf1) phosphorylation deregulates its activity and lowers activation threshold, leading to obesity in mice
- Author
-
Xiongjie Jin, Nahid F. Mivechi, Demetrius Moskophidis, and Aijun Qiao
- Subjects
0301 basic medicine ,fungi ,Wild type ,Stimulation ,Cell Biology ,Biology ,medicine.disease ,Biochemistry ,Serine ,03 medical and health sciences ,030104 developmental biology ,Insulin resistance ,In vivo ,Mutant protein ,medicine ,Phosphorylation ,HSF1 ,Molecular Biology - Abstract
Heat shock factor 1 (Hsf1) is transcriptionally activated following exposure of mammalian cells to environmental insults and pathological conditions. The function of Hsf1 as a protector of cells and organisms towards a range of stress stimuli has been well-documented. However, how different Hsf1 post-transcriptional modifications affect its function in vivo is not understood. Hsf1 transcriptional activity is in part regulated by phosphorylation. Hsf1 phosphorylation of amino acid residues at S303 and S307 has been shown to repress Hsf1 transcriptional activity under normal physiological growth conditions. In this study, we used a knock-in mouse model where serine residues S303 and S307 were mutated to alanine (S303A/S307A) in order to reveal the relevance of these phosphorylation sites on Hsf1 activity in vitro and in vivo. Our results indicate that Hsf1303A/307A protein becomes more stable, but remains mostly cytoplasmic under normal physiological growth conditions. However, in contrast to wild type, the Hsf1303A/307A protein exhibits reduced threshold of activation following exposure of cells to a very mild heat stress, or when mice are placed under the conditions of nutrient stimulation. The in vivo consequence of the Hsf1303A/307A mutant protein expression results in an age-dependent obesity, fatty liver disease and insulin resistance. Taken together, these data point to the importance of maintaining dynamic regulation of Hsf1 responses including resolution of Hsf1 activation to restore repression of Hsf1 through phosphorylation of S303 and/or S307. Specifically, we show that the inability to phosphorylate Hsf1 at these amino acid residues can play a role in the development of age-dependent metabolic diseases, such as obesity and insulin resistance.
- Published
- 2017
18. Therapeutic Inducers of the HSP70/HSP110 Protect Mice Against Traumatic Brain Injury
- Author
-
Donald E. Kimbler, Krishnan M. Dhandapani, Junfeng Pang, Demetrius Moskophidis, Nathan Yanasak, Nahid F. Mivechi, Binnur Eroglu, and Justin J Choi
- Subjects
Programmed cell death ,Traumatic brain injury ,Immunoblotting ,HSP72 Heat-Shock Proteins ,Pharmacology ,Biochemistry ,Article ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Piperidines ,Heat shock protein ,Oximes ,Medicine ,Animals ,Inducer ,Fear conditioning ,HSP110 Heat-Shock Proteins ,Pathological ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,business.industry ,Brain ,medicine.disease ,Immunohistochemistry ,Triterpenes ,Hsp70 ,nervous system diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,chemistry ,nervous system ,Gene Expression Regulation ,Celastrol ,Brain Injuries ,Tumor Suppressor Protein p53 ,business ,Pentacyclic Triterpenes ,Reactive Oxygen Species ,Neuroscience ,Multiplex Polymerase Chain Reaction - Abstract
Traumatic brain injury (TBI) induces severe harm and disability in many accident victims and combat-related activities. The heat-shock proteins Hsp70/Hsp110 protect cells against death and ischemic damage. In this study, we used mice deficient in Hsp110 or Hsp70 to examine their potential requirement following TBI. Data indicate that loss of Hsp110 or Hsp70 increases brain injury and death of neurons. One of the mechanisms underlying the increased cell death observed in the absence of Hsp110 and Hsp70 following TBI is the increased expression of reactive oxygen species-induced p53 target genes Pig1, Pig8, and Pig12. To examine whether drugs that increase the levels of Hsp70/Hsp110 can protect cells against TBI, we subjected mice to TBI and administered Celastrol or BGP-15. In contrast to Hsp110- or Hsp70i-deficient mice that were not protected following TBI and Celastrol treatment, there was a significant improvement of wild-type mice following administration of these drugs during the first week following TBI. In addition, assessment of neurological injury shows significant improvement in contextual and cued fear conditioning tests and beam balance in wild-type mice that were treated with Celastrol or BGP-15 following TBI compared to TBI-treated mice. These studies indicate a significant role of Hsp70/Hsp110 in neuronal survival following TBI and the beneficial effects of Hsp70/Hsp110 inducers toward reducing the pathological consequences of TBI. Our data indicate that loss of Hsp110 or Hsp70 in mice increases brain injury following TBI. (a) One of the mechanisms underlying the increased cell death observed in the absence of these Hsps following TBI is the increased expression of ROS-induced p53 target genes known as Pigs. In addition, (b) using drugs (Celastrol or BGP-15) to increase Hsp70/Hsp110 levels protect cells against TBI, suggesting the beneficial effects of Hsp70/Hsp110 inducers to reduce the pathological consequences of TBI.
- Published
- 2014
19. Insights into Regulation and Function of the Major Stress-Induced hsp70 Molecular Chaperone In Vivo: Analysis of Mice with Targeted Gene Disruption of the hsp70.1 orhsp70.3 Gene
- Author
-
Demetrius Moskophidis, Lei Huang, and Nahid F. Mivechi
- Subjects
TBX1 ,Hot Temperature ,Time Factors ,Fever ,Genotype ,Transcription, Genetic ,Cell Survival ,Blotting, Western ,Protozoan Proteins ,Apoptosis ,Bone Marrow Cells ,Cytochrome c Group ,Biology ,Evolution, Molecular ,Mice ,Stress, Physiological ,In vivo ,Transcription (biology) ,Mammalian Genetic Models with Minimal or Complex Phenotypes ,medicine ,Animals ,HSP70 Heat-Shock Proteins ,Tissue Distribution ,Molecular Biology ,Gene ,Alleles ,Gene Library ,Models, Genetic ,Temperature ,Cell Biology ,beta-Galactosidase ,Molecular biology ,Epithelium ,Hsp70 ,Mice, Inbred C57BL ,Blot ,medicine.anatomical_structure ,Caspases ,Mutagenesis, Site-Directed ,Function (biology) ,Molecular Chaperones - Abstract
The murine hsp70 gene family includes the evolutionarily conserved hsp70.1 and hsp70.3 genes, which are the major proteins induced by heat and other stress stimuli. hsp70.1 and hsp70.3 encode identical proteins which protect cells and facilitate their recovery from stress-induced damage. While the hsp70 gene family has been widely studied and the roles of the proteins it encodes as molecular chaperones in a range of human pathologies are appreciated, little is known about the developmental regulation of hsp70.1 and hsp70.3 expression and the in vivo biological function of their products. To directly study the physiological role of these proteins in vivo, we have generated mice deficient in heat shock protein 70 (hsp70) by replacing the hsp70.1 or hsp70.3 gene with an in-frame beta-galactosidase sequence. We report here that the expression of hsp70.1 and hsp70.3 is developmentally regulated at the transcriptional level, and an overlapping expression pattern for both genes is observed during embryo development and in the tissues of adult mice. hsp70.1-/- or hsp70.3-/- mice are viable and fertile, with no obvious morphological abnormalities. In late embryonic stage and adult mice, both genes are expressed constitutively in tissues exposed directly to the environment (the epidermis and cornea) and in certain internal organs (the epithelium of the tongue, esophagus, and forestomach, and the kidney, bladder, and hippocampus). Exposure of mice to thermal stress results in the rapid induction and expression of hsp70, especially in organs not constitutively expressing hsp70 (the liver, pancreas, heart, lung, adrenal cortex, and intestine). Despite functional compensation in the single-gene-deficient mice by the intact homologous gene (i.e., hsp70.3 in hsp70.1-/- mice and vice versa), a marked reduction in hsp70 protein expression was observed in tissues under both normal and heat stress conditions. At the cellular level, inactivation of hsp70.1 or hsp70.3 resulted in deficient maintenance of acquired thermotolerance and increased sensitivity to heat stress-induced apoptosis. The additive or synergistic effects exhibited by coexpression of both hsp70 genes, and the evolutionary significance of the presence of both hsp70 genes, is hence underlined.
- Published
- 2001
20. Critical Role for Alpha/Beta and Gamma Interferons in Persistence of Lymphocytic Choriomeningitis Virus by Clonal Exhaustion of Cytotoxic T Cells
- Author
-
Lei Huang, Shenghua Zhou, Rong Ou, and Demetrius Moskophidis
- Subjects
CD4-Positive T-Lymphocytes ,viruses ,Immunology ,Alpha interferon ,Receptor, Interferon alpha-beta ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,Microbiology ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Neutralization Tests ,Interferon ,Virology ,Virus latency ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Cytotoxic T cell ,Interferon gamma ,Receptors, Interferon ,Mice, Knockout ,Interferon-alpha ,Membrane Proteins ,Interferon-beta ,medicine.disease ,Virus Latency ,Insect Science ,Acute Disease ,Pathogenesis and Immunity ,T-Lymphocytes, Cytotoxic ,medicine.drug - Abstract
Under conditions of high antigenic load during infection with invasive lymphocytic choriomeningitis virus (LCMV) strains, virus can persist by selective clonal exhaustion of antigen-specific CD8+T cells. In this work we studied the down-regulation of the virus-specific CD8+-T-cell response during a persistent infection of adult mice, with particular emphasis on the contribution of the interferon response in promoting host defense. Studies were conducted by infecting mice deficient in receptors for type I (alpha/beta interferon [IFN-α/β]), type II (IFN-γ), and both type I and II IFNs with LCMV isolates that vary in their capacity to induce T-cell exhaustion. The main conclusions of this study are as follows. (i) IFNs play a critical role in LCMV infection by reducing viral loads in the initial stages of infection and thus modifying both the extent of CD8+-T-cell exhaustion and the course of infection. The importance of IFNs in this context varies with the biological properties of the LCMV strain. (ii) An inverse correlation exists between antigen persistence and responsiveness of virus-specific CD8+T cells. This results in distinct programs of activation or tolerance (functional unresponsiveness and/or physical elimination of antigen-specific cells) during acute and chronic virus infections, respectively. (iii) A successful immune response associated with definitive viral clearance requires an appropriate balance between cellular and humoral components of the immune system. We discuss the role of IFNs in influencing virus-specific T cells that determine the outcome of persistent infections.
- Published
- 2001
21. Viral Escape by Selection of Cytotoxic T Cell–Resistant Variants in Influenza a Virus Pneumonia
- Author
-
Demetrius Moskophidis, Rong Ou, Graeme E. Price, Lei Huang, and Hong Jiang
- Subjects
Cytotoxicity, Immunologic ,influenza viral pneumonia ,viruses ,Epitopes, T-Lymphocyte ,medicine.disease_cause ,viral persistence ,Mice ,Virus latency ,Influenza A virus ,Immunology and Allergy ,Antigen Presentation ,biology ,Viral Core Proteins ,RNA-Binding Proteins ,Nucleocapsid Proteins ,Antigenic Variation ,Virus Latency ,T cell receptor transgenic mice ,Original Article ,Immunocompetence ,Cell Division ,Pneumonia, Viral ,Immunology ,Orthomyxoviridae ,CD8+ CTL escape variants ,Hemagglutinin (influenza) ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Virus ,Antigenic drift ,Cell Line ,Dogs ,Influenza, Human ,medicine ,Animals ,Humans ,influenza A virus ,Homeodomain Proteins ,biology.organism_classification ,medicine.disease ,Virology ,Peptide Fragments ,Disease Models, Animal ,CTL ,Nucleoproteins ,biology.protein ,Peptides ,Neuraminidase ,T-Lymphocytes, Cytotoxic - Abstract
Antigenic variation is a strategy exploited by influenza viruses to promote survival in the face of the host adaptive immune response and constitutes a major obstacle to efficient vaccine development. Thus, variation in the surface glycoproteins hemagglutinin and neuraminidase is reflected by changes in susceptibility to antibody neutralization. This has led to the current view that antibody-mediated selection of influenza A viruses constitutes the basis for annual influenza epidemics and periodic pandemics. However, infection with this virus elicits a vigorous protective CD8+ cytotoxic T lymphocyte (CTL) response, suggesting that CD8+ CTLs might exert selection pressure on the virus. Studies with influenza A virus–infected transgenic mice bearing a T cell receptor (TCR) specific for viral nucleoprotein reveal that virus reemergence and persistence occurs weeks after the acute infection has apparently been controlled. The persisting virus is no longer recognized by CTLs, indicating that amino acid changes in the major viral nucleoprotein CTL epitope can be rapidly accumulated in vivo. These mutations lead to a total or partial loss of recognition by polyclonal CTLs by affecting presentation of viral peptide by class I major histocompatibility complex (MHC) molecules, or by interfering with TCR recognition of the mutant peptide–MHC complex. These data illustrate the distinct features of pulmonary immunity in selection of CTL escape variants. The likelihood of emergence and the biological impact of CTL escape variants on the clinical outcome of influenza pneumonia in an immunocompetent host, which is relevant for the design of preventive vaccines against this and other respiratory viral infections, are discussed.
- Published
- 2000
22. The Role of Alpha/Beta and Gamma Interferons in Development of Immunity to Influenza A Virus in Mice
- Author
-
Graeme E. Price, Demetrius Moskophidis, and Anna Gaszewska-Mastarlarz
- Subjects
Secondary infection ,Pneumonia, Viral ,Immunology ,Receptor, Interferon alpha-beta ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,Interferon-gamma ,Mice ,Dogs ,Immune system ,Orthomyxoviridae Infections ,Interferon ,Virology ,medicine ,Influenza A virus ,Animals ,Antibody-dependent enhancement ,Lung ,Receptors, Interferon ,Mice, Knockout ,Interferon-alpha ,Membrane Proteins ,Interferon-beta ,Acquired immune system ,Insect Science ,Tissue tropism ,Pathogenesis and Immunity ,Disease Susceptibility ,T-Lymphocytes, Cytotoxic ,medicine.drug - Abstract
During influenza virus infection innate and adaptive immune defenses are activated to eliminate the virus and thereby bring about recovery from illness. Both arms of the adaptive immune system, antibody neutralization of free virus and termination of intracellular virus replication by antiviral cytotoxic T cells (CTLs), play pivotal roles in virus elimination and protection from disease. Innate cytokine responses, such as alpha/beta interferon (IFN-α/β) or IFN-γ, can have roles in determining the rate of virus replication in the initial stages of infection and in shaping the initial inflammatory and downstream adaptive immune responses. The effect of these cytokines on the replication of pneumotropic influenza A virus in the respiratory tract and in the regulation of adaptive antiviral immunity was examined after intranasal infection of mice with null mutations in receptors for IFN-α/β, IFN-γ, and both IFNs. Virus titers in the lungs of mice unable to respond to IFNs were not significantly different from congenic controls for both primary and secondary infection. Likewise the mice were comparably susceptible to X31 (H3N2) influenza virus infection. No significant disruption to the development of normal antiviral CTL or antibody responses was observed. In contrast, mice bearing the disrupted IFN-α/β receptor exhibited accelerated kinetics and significantly higher levels of neutralizing antibody activity during primary or secondary heterosubtypic influenza virus infection. Thus, these observations reveal no significant contribution for IFN-controlled pathways in shaping acute or memory T-cell responses to pneumotropic influenza virus infection but do indicate some role for IFN-α/β in the regulation of antibody responses. Recognizing the pivotal role of CTLs and antibody in virus clearance, it is reasonable to assume a redundancy in IFN-mediated antiviral effects in pulmonary influenza. However, IFN-α/β seems to be a valid factor in determining tissue tropism and replicative rates of highly virulent influenza virus strains as reported previously by others, and this aspect is discussed here.
- Published
- 2000
23. Contribution of Virus-specific CD8+ Cytotoxic T Cells to Virus Clearance or Pathologic Manifestations of Influenza Virus Infection in a T Cell Receptor Transgenic Mouse Model
- Author
-
Demetrius Moskophidis and Dimitris Kioussis
- Subjects
Cytotoxicity, Immunologic ,influenza viral pneumonia ,viruses ,Receptors, Antigen, T-Cell, alpha-beta ,CD8+ cytotoxic T lymphocytes ,T cell ,Immunology ,Orthomyxoviridae ,Mice, Transgenic ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Biology ,medicine.disease_cause ,Virus ,Mice ,Orthomyxoviridae Infections ,Antigen ,Influenza A virus ,medicine ,Animals ,influenza A virus ,Immunology and Allergy ,Cytotoxic T cell ,interferon γ ,Articles ,T cell receptor–transgenic mice ,biology.organism_classification ,Virology ,CTL ,medicine.anatomical_structure ,CD8 - Abstract
The ability of influenza virus to evade immune surveillance by neutralizing antibodies (Abs) directed against its variable surface antigens provides a challenge to the development of effective vaccines. CD8+ cytotoxic T lymphocytes (CTLs) restricted by class I major histocompatibility complex molecules are important in establishing immunity to influenza virus because they recognize internal viral proteins which are conserved between multiple viral strains. In contrast, protective Abs are strain-specific. However, the precise role of effector CD8+ CTLs in protection from influenza virus infection, critical for understanding disease pathogenesis, has not been well defined. In transgenic mice with a very high frequency of antiinfluenza CTL precursors, but without protective Abs, CD8+ CTLs conferred protection against low dose viral challenge, but exacerbated viral pathology and caused mortality at high viral dose. The data suggest a dual role for CD8+ CTLs against influenza, which may present a challenge to the development of effective CTL vaccines. Effector mechanisms used by CD8+ CTLs in orchestrating clearance of virus and recovery from experimental influenza infection, or potentiation of lethal pathology, are discussed.
- Published
- 1998
24. Immunobiology of cytotoxic T-cell resistant virus variants: studies on lymphocytic choriomeningitis virus (LCMV)
- Author
-
Demetrius Moskophidis and Rolf M. Zinkernagel
- Subjects
biology ,Immunology ,chemical and pharmacologic phenomena ,Immunodominance ,Major histocompatibility complex ,Lymphocytic choriomeningitis ,medicine.disease ,Virology ,Virus ,Epitope ,CTL ,Antigen ,biology.protein ,medicine ,Cytotoxic T cell - Abstract
Replication of the genetically variable lymphocytic choriomeningitis virus (LCMV) gives rise to a pool of variant viruses. Under the selection pressure exerted by a strong but narrow repertoire of antiviral cytotoxic T-cells (CTL) i.e. monoclonal or polyclonal monoepitope, variant viruses emerge that contain point mutations in the nucleotide sequence encoding antigenic CTL epitopes; these variants can be selected in both infected mice and cell cultures. These mutations permit infected cells to escape CTL recognition by altering the ability of the mutant peptides to bind MHC class-I-molecules or by interfering with the ability of T-cell receptors to interact with the mutant peptide/MHC complex. Because viral infections often trigger a polyclonal repertoire of antiviral CTL to multiple epitopes, the likelihood of selection of CTL resistant variants is probably low, but not impossible. Our empirical observations suggest that antigenic variations, even if they only occur in a part of the available CTL epitope, may exert significant effects on the subtle biological equilibrium established between virus and host immune system. This can reduce immunological control of the pathogen population, and so permit persistence of viral infection and promote disease progression.
- Published
- 1996
25. Immunobiology of cytotoxic T-cell escape mutants of lymphocytic choriomeningitis virus
- Author
-
R M Zinkernagel and Demetrius Moskophidis
- Subjects
Cytotoxicity, Immunologic ,Molecular Sequence Data ,Immunology ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,Microbiology ,Virus ,Epitope ,Epitopes ,Mice ,Immune system ,Virology ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Point Mutation ,Cytotoxic T cell ,Amino Acid Sequence ,Mice, Inbred BALB C ,Base Sequence ,medicine.disease ,Nucleoprotein ,Mice, Inbred C57BL ,CTL ,Insect Science ,DNA, Viral ,CD8 ,T-Lymphocytes, Cytotoxic ,Research Article - Abstract
Infection with virus variants exhibiting changes in the peptide sequences defining immunodominant determinants that abolish recognition by antiviral cytotoxic T cells (CTL) presents a considerable challenge to the antiviral T-cell immune system and may enable some viruses to persist in hosts. The potential importance of such variants with respect to mechanisms of viral persistence and disease pathogenesis was assessed by infecting adult mice with variants of lymphocytic choriomeningitis virus (LCMV) strain WE. These variants were selected in vivo or in vitro for resistance to lysis by CD8+ H-2b-restricted antiviral CTL. The majority of anti-LCMV CTL in infected H-2b mice recognize epitopes defined by residues 32 to 42 and 275 to 289 (epitopes 32-42 and 275-289) of the LCMV glycoprotein or 397 to 407 of the viral nucleoprotein. The 8.7 variant exhibits a change in the epitope 32-42 (Val-35-->Leu), and variant CL1.2 exhibits a change in the epitope 275-289 (Asn-280-->Asp) of the wild-type LCMV-WE. The double-mutated 8.7-B23 variant had the variation of 8.7 and an additional change located in the epitope 275-289 (Asn-280-->Ser). The 8.7 variant peptide with unchanged anchor positions bound efficiently to H-2Db and H-2Kb molecules but induced only a very weak CTL response. CTL epitope 275-289 of CL1.2 and 8.7-B23 altered at predicted anchor residues were unable to bind Db molecules and were also not recognized by antiviral CTL. Infection of C57BL/6 mice (H-2b) with the variants exhibiting mutations of one of the CTL epitopes, i.e., 8.7 or CL1.2, induced CTL responses specific for the unmutated epitopes comparable with those induced by infection with WE, and these responses were sufficient to eliminate virus from the host. In contrast, infection with the double-mutated variant 8.7-B23 induced CTL activity that was reduced by a factor of about 50-fold compared with wild-type LCMV. Consequently, high doses (10(7) PFU intravenously) of this virus were eliminated slowly and only by about day 100 after infection. 8.7-B23 failed to cause lethal lymphocytic choriomeningitis after intracerebral infection with a dose of > 10(4) PFU in C57BL/6 mice (but not in mice of nonselecting H-2d haplotype); with the other variants or wild-type LCMV, doses greater than 10(6) to 10(7) PFU were necessary to avoid lethal choriomeningitis.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1995
26. Inactivation of heat shock factor Hsf4 induces cellular senescence and suppresses tumorigenesis in vivo
- Author
-
Binnur Eroglu, Yukihiro Yamaguchi, Wonkyoung Cho, Xiongjie Jin, Nahid F. Mivechi, and Demetrius Moskophidis
- Subjects
Senescence ,Cancer Research ,Transgene ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Article ,Mice ,Heat Shock Transcription Factors ,In vivo ,medicine ,Animals ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Kinase ,Heat shock factor ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,Oncology ,Cancer research ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Cell aging ,Transcription Factors - Abstract
Studies suggest that Hsf4 expression correlates with its role in cell growth and differentiation. However, the role of Hsf4 in tumorigenesis in vivo remains unexplored. In this article, we provide evidence that absence of the Hsf4 gene suppresses evolution of spontaneous tumors arising in p53- or Arf-deficient mice. Furthermore, deletion of hsf4 alters the tumor spectrum by significantly inhibiting development of lymphomas that are normally observed in the majority of mice lacking p53 or Arf tumor suppressor genes. Using mouse embryo fibroblasts deficient in the hsf4 gene, we have found that these cells exhibit reduced proliferation that is associated with induction of senescence and senescence-associated β-galactosidase (SA-β-gal). Cellular senescence in hsf4-deficient cells is associated with the increased expression of the cyclin-dependent kinase inhibitors, p21 and p27 proteins. Consistent with the cellular senescence observed in vitro, specific normal tissues of hsf4−/− mice and tumors that arose in mice deficient in both hsf4 and p53 genes exhibit increased SA-β-gal activity and elevated levels of p27 compared with wild-type mice. These results suggest that hsf4 deletion-induced senescence is also present in vivo. Our results therefore indicate that Hsf4 is involved in modulation of cellular senescence, which can be exploited during cancer therapy. Mol Cancer Res; 10(4); 523–34. ©2012 AACR.
- Published
- 2012
27. Resistance of lymphocytic choriomeningitis virus to alpha/beta interferon and to gamma interferon
- Author
-
Ion Gresser, Elisabeth Laine, Manuel Battegay, Rolf M. Zinkernagel, Marie Anne Bruendler, and Demetrius Moskophidis
- Subjects
Immunology ,Alpha interferon ,Biology ,Virus Replication ,Lymphocytic choriomeningitis ,Microbiology ,Virus ,Cell Line ,Interferon-gamma ,Mice ,Interferon ,Virology ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Interferon gamma ,Beta (finance) ,Interferon alfa ,Arenavirus ,Macrophages ,Interferon-alpha ,Drug Resistance, Microbial ,Interferon-beta ,Fibroblasts ,medicine.disease ,biology.organism_classification ,Mice, Inbred C57BL ,Insect Science ,Spleen ,Research Article ,medicine.drug - Abstract
The susceptibility to alpha/beta interferon (IFN-alpha/beta) or to gamma interferon (IFN-gamma) of various lymphocytic choriomeningitis virus (LCMV) strains was evaluated in C57BL/6 mice and in various cell lines. Anti-IFN-gamma treatment in vivo revealed that the LCMV strains Armstrong, Aggressive, and WE were most susceptible to IFN-gamma whereas Traub, Cl 13-Armstrong, and Docile were resistant. The same pattern of susceptibility to recombinant IFN-gamma was observed in vitro. In vivo treatment with anti-IFN-alpha/beta showed a sizeable increase in replication of Aggressive, Armstrong, and WE; effects were less pronounced for Docile, Cl 13-Armstrong, or Traub. Correspondingly, WE, Armstrong, and Aggressive were all relatively sensitive to purified IFN-alpha/beta in vitro, and Cl 13-Armstrong, Docile, and Traub were more resistant. Overall, there was a good correlation between the capacity of LCMV strains to establish a persistent infection in adult immunocompetent mice and their relative resistance to IFN-gamma and IFN-alpha/beta.
- Published
- 1994
28. Targeted Deletion of Hsf1, 2, and 4 Genes in Mice
- Author
-
Binnur Eroglu, Xiongjie Jin, Demetrius Moskophidis, and Nahid F. Mivechi
- Subjects
0301 basic medicine ,Male ,Genetic Vectors ,Biology ,DNA-binding protein ,Article ,Cell Line ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Heat Shock Transcription Factors ,Heat shock protein ,Animals ,Heat shock ,HSF1 ,Transcription factor ,Heat-Shock Proteins ,Sequence Deletion ,Mice, Knockout ,Base Sequence ,Promoter ,Molecular biology ,Heat shock factor ,DNA-Binding Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Knockout mouse ,Gene Deletion ,Heat-Shock Response ,Transcription Factors - Abstract
Heat shock transcription factors (Hsfs) regulate transcription of heat shock proteins as well as other genes whose promoters contain heat shock elements (HSEs). There are at least five Hsfs in mammalian cells, Hsf1, Hsf2, Hsf3, Hsf4, and Hsfy (Wu, Annu Rev Cell Dev Biol 11:441-469, 1995; Morimoto, Genes Dev 12:3788-3796, 1998; Tessari et al., Mol Hum Repord 4:253-258, 2004; Fujimoto et al., Mol Biol Cell 21:106-116, 2010; Nakai et al., Mol Cell Biol 17:469-481, 1997; Sarge et al., Genes Dev 5:1902-1911, 1991). To understand the physiological roles of Hsf1, Hsf2, and Hsf4 in vivo, we generated knockout mouse lines for these factors (Zhang et al., J Cell Biochem 86:376-393, 2002; Wang et al., Genesis 36:48-61, 2003; Min et al., Genesis 40:205-217, 2004). Numbers of other laboratories have also generated Hsf1 (Xiao et al., EMBO J 18:5943-5952, 1999; Sugahara et al., Hear Res 182:88-96, 2003), Hsf2 (McMillan et al., Mol Cell Biol 22:8005-8014, 2002; Kallio et al., EMBO J 21:2591-2601, 2002), and Hsf4 (Fujimoto et al., EMBO J 23:4297-4306, 2004) knockout mouse models. In this chapter, we describe the design of the targeting vectors, the plasmids used, and the successful generation of mice lacking the individual genes. We also briefly describe what we have learned about the physiological functions of these genes in vivo.
- Published
- 2011
29. Peripheral clonal deletion of antiviral memory CD8+ T cells
- Author
-
Rolf M. Zinkernagel, Demetrius Moskophidis, and Elisabeth Laine
- Subjects
Adoptive cell transfer ,CD8 Antigens ,Immunology ,Clonal Deletion ,Mice, Transgenic ,T lymphocyte ,Lymphocytic Choriomeningitis ,Biology ,Immunotherapy, Adoptive ,Virology ,Clonal deletion ,Mice, Inbred C57BL ,Mice ,Interleukin 21 ,Immune system ,Animals ,Lymphocytic choriomeningitis virus ,Immunology and Allergy ,Cytotoxic T cell ,Antigen-presenting cell ,Immunologic Memory ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
Antiviral cytotoxic memory CD8+ T cells adoptively transferred to mice which are persistently infected with lymphocytic choriomeningitis virus WE or DOCILE initially proliferated extensively; they either caused the death of the recipient or, alternatively, disappeared within a few days. Apparently, the complete and coordinated induction and stimulation by widely distributed viral antigen caused these memory T cells to die before virus had been eliminated from the host. Thus memory T cells are as susceptible to peripheral exhaustion/deletion as unprimed T cells. These results indicate possible limitations of exclusively CD8+ T cell-mediated adoptive immunotherapy against viral infections or tumors.
- Published
- 1993
30. Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells
- Author
-
Hanspeter Pircher, Franziska Lechner, Demetrius Moskophidis, and Rolf M. Zinkernagel
- Subjects
Cellular immunity ,CD8 Antigens ,viruses ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,Immunotherapy, Adoptive ,Virus ,Immunophenotyping ,Mice ,Immune system ,Immunity ,Immune Tolerance ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Cytotoxic T cell ,Tropism ,Multidisciplinary ,Arenavirus ,biology.organism_classification ,medicine.disease ,Virology ,Mice, Inbred C57BL ,Immunology ,Immunocompetence ,Spleen ,T-Lymphocytes, Cytotoxic - Abstract
Viruses that are non- or poorly cytopathic have developed various strategies to avoid elimination by the immune system and to persist in the host. Acute infection of adult mice with the noncytopathic lymphocytic choriomeningitis virus (LCMV) normally induces a protective cytotoxic T-cell response that also causes immunopathology. But some LCMV strains (such as DOCILE (LCMV-D) or Cl-13 Armstrong (Cl-13)) derived from virus carrier mice tend to persist after acute infection of adult mice without causing lethal immunopathological disease. Tendency to persist correlates with tropism, rapidity of virus spread and virus mutations. We report here that these LCMV isolates may persist because they induce most of the specific antiviral CD8+ cytotoxic T cells so completely that they all disappear within a few days and therefore neither eliminate the virus nor cause lethal immunopathology. The results illustrate that partially and sequentially induced (protective) immunity or complete exhaustion of T-cell immunity (high zone tolerance) are quantitatively different points on the scale of immunity; some viruses exploit the latter possibility to persist in an immunocompetent host.
- Published
- 1993
31. Lower receptor avidity required for thymic clonal deletion than for effector T-cell function
- Author
-
Urs Hoffmann Rohrer, Rolf M. Zinkernagel, Demetrius Moskophidis, Hans Hengartner, and Hanspeter Pircher
- Subjects
T cell ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Thymus Gland ,Biology ,Lymphocytic choriomeningitis ,Epitope ,Clonal deletion ,Immune tolerance ,Epitopes ,Mice ,Immune Tolerance ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Avidity ,Amino Acid Sequence ,Antigens, Viral ,Multidisciplinary ,Base Sequence ,T-cell receptor ,medicine.disease ,Virology ,Molecular biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Mutation ,Clone (B-cell biology) ,T-Lymphocytes, Cytotoxic - Abstract
CLONAL deletion in the thymus plays a major part in T-cell tolerance to self antigens1–3. But the mechanism of negative selection, its fine specificity and the threshold of affinity and avidity remains unknown. We have now examined these aspects of negative selection with mice expressing a transgenic T-cell receptor with specificity for lymphocytic choriomeningitis virus (LCMV) glycoprotein in association with the class I H–2Db molecule. These mice were rendered tolerant to LCMV by neonatal infection with mutant LCMVs hearing point mutations in the T-cell epitope recognized by the transgenic T-cell receptor (ref. 4). Variant LCMVs were also tested for their ability to elicit antiviral responses in transgenic mice in vivo and in vitro. Comparison in vivo revealed that a low-avidity receptor interaction, which was unable to induce effector T cells in the periphery, was still sufficient for clonal deletion in the thymus.
- Published
- 1991
32. Genomic and biological characterization of aggressive and docile strains of lymphocytic choriomeningitis virus rescued from a plasmid-based reverse-genetics system
- Author
-
Shuiyun Lan, Rong Ou, Hong Jiang, Juan Carlos de la Torre, Demetrius Moskophidis, Graeme E. Price, and Minjie Chen
- Subjects
viruses ,Virulence ,Disease ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis ,Virus ,Article ,Cell Line ,Rodent Diseases ,Mice ,Plasmid ,Virology ,Cricetinae ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Genetics ,Arenavirus ,biology ,Sequence Analysis, RNA ,virus diseases ,biology.organism_classification ,medicine.disease ,Phenotype ,Reverse genetics ,Mice, Inbred C57BL ,Host-Pathogen Interactions ,RNA, Viral ,Reassortant Viruses ,Plasmids - Abstract
Arenaviruses include several causative agents of haemorrhagic fever disease in humans. In addition, the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a superb model for the study of virus–host interactions, including the basis of viral persistence and associated diseases. There is little understanding about the molecular mechanisms concerning the regulation and specific role of viral proteins in modulating arenavirus–host cell interactions either associated with an acute or persistent infection, and associated disease. Here, we report the genomic and biological characterization of LCMV strains ‘Docile’ (persistent) and ‘Aggressive’ (not persistent) recovered from cloned cDNA via reverse genetics. Our results confirmed that the cloned viruses accurately recreated thein vivophenotypes associated with the corresponding natural Docile and Aggressive viral isolates. In addition, we provide evidence that the ability of the Docile strain to persist is determined by the nature of both S and L RNA segments. Thus, our findings provide the foundation for studies aimed at gaining a detailed understanding of viral determinants of LCMV persistence in its natural host, which may aid in the development of vaccines to prevent or treat the diseases caused by arenaviruses in humans.
- Published
- 2008
33. Unique contribution of heat shock transcription factor 4 in ocular lens development and fiber cell differentiation
- Author
-
Demetrius Moskophidis, Nahid F. Mivechi, Jin Na Min, and Yan Zhang
- Subjects
Biology ,Mice ,Endocrinology ,Cataracts ,Heat Shock Transcription Factors ,Heat shock protein ,Lens, Crystalline ,Genetics ,medicine ,Animals ,Transcription factor ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,medicine.disease ,Molecular biology ,Mice, Mutant Strains ,Cell biology ,Heat shock factor ,DNA-Binding Proteins ,medicine.anatomical_structure ,Phenotype ,Fiber cell ,Membrane protein ,Lens (anatomy) ,Gene Targeting ,Eye development ,Transcription Factors - Abstract
Mammalian ocular lens development results via a differentiation program that is highly regulated by tissue-specific transcription factors. Central to this is the terminal differentiation of fiber cells, which develop from epithelial cells on the anterior surface of the lens, accompanied by a change in cell shape and expression of structural proteins (such as membrane proteins MP19, MIP26, connexin 43, 46, and 50, cytoskeletal proteins CP49, CP115, and α, β, and γ crystallins), creating a transparent, refractive index gradient in the lens. Mutations in genes controlling eye development and in lens structural protein genes are associated with multiple ocular developmental disorders, including cataracts and other opacities of the lens. Here we show that heat shock transcription factor 4 (HSF4) expression in the developing lens is required for correct lens development and that inactivation of hsf4 leads to early postnatal cataract formation with primary effects specific to terminal fiber cell differentiation. These data suggest that HSF4 acts as a critical transcription factor for lens-specific target gene expression, in particular regulating the small 25 kDa heat shock protein that acts as a modifier for lens opacity and cataract development. Thus, HSF4 fulfills a central role in controlling spatial and temporal expression of genes critical for correct development and function of the lens. genesis 40:205–217, 2004. © 2004 Wiley-Liss, Inc.
- Published
- 2004
34. TCR affinity and negative regulation limit autoimmunity
- Author
-
Awen Gallimore, Kaisa Holmberg, Linh T. Nguyen, Elissa K. Deenick, Demetrius Moskophidis, Matthew A. Gronski, Hee O Kim, Bernhard Odermatt, Alisha R. Elford, Josef M. Penninger, Nicholas R. J. Gascoigne, Jonathan M. Boulter, and Pamela S. Ohashi
- Subjects
Receptors, Antigen, T-Cell ,Autoimmunity ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Ligands ,General Biochemistry, Genetics and Molecular Biology ,Diabetes Mellitus, Experimental ,Mice ,Immune system ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,Receptor ,Cell Proliferation ,Autoimmune disease ,T-cell receptor ,Molecular Mimicry ,Models, Immunological ,General Medicine ,medicine.disease ,Ligand (biochemistry) ,Flow Cytometry ,Immunohistochemistry ,Chromium Radioisotopes ,Mice, Inbred C57BL ,Molecular mimicry ,Immunology ,T-Lymphocytes, Cytotoxic - Abstract
Autoimmune diseases are often mediated by self-reactive T cells, which must be activated to cause immunopathology. One mechanism, known as molecular mimicry, proposes that self-reactive T cells may be activated by pathogens expressing crossreactive ligands1,2,3. Here we have developed a model to investigate how the affinity of the T-cell receptor (TCR) for the activating agent influences autoimmunity. Our model shows that an approximately fivefold difference in the TCR affinity for the activating ligand results in a 50% reduction in the incidence of autoimmunity. A reduction in TCR-ligand affinity to approximately 20 times lower than normal does not induce autoimmunity despite the unexpected induction of cytotoxic T lymphocytes (CTLs) and insulitis. Furthermore, in the absence of a key negative regulatory molecule, Cbl-b4,5, 100% of mice develop autoimmunity upon infection with viruses encoding the lower-affinity ligand. Therefore, autoimmune disease is sensitive both to the affinity of the activating ligand and to normal mechanisms that negatively regulate the immune response.
- Published
- 2004
35. Targeted disruption of hsf1 leads to lack of thermotolerance and defines tissue-specific regulation for stress-inducible Hsp molecular chaperones
- Author
-
Yan Zhang, Jing Zhang, Demetrius Moskophidis, Lei Huang, and Nahid F. Mivechi
- Subjects
Hot Temperature ,Apoptosis ,Bone Marrow Cells ,Biology ,In Vitro Techniques ,Biochemistry ,Transactivation ,Mice ,Heat Shock Transcription Factors ,Gene knockin ,Heat shock protein ,Animals ,HSP70 Heat-Shock Proteins ,Heat shock ,HSF1 ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Heat-Shock Proteins ,Regulation of gene expression ,fungi ,Cell Biology ,Fibroblasts ,Hsp70 ,Cell biology ,DNA-Binding Proteins ,Gene Expression Regulation ,Organ Specificity ,Heat-Shock Response ,Molecular Chaperones ,Transcription Factors - Abstract
The rapid synthesis of heat shock proteins (Hsps) in cells subjected to environmental challenge is controlled by heat shock transcription factor-1 (Hsf1). Regulation of Hsps by Hsf1 is highly complex and, in the whole organism, remains largely unexplored. In this study, we have used mouse embryo fibroblasts and bone marrow progenitor cells from hsf1-/- mice as well as hsp70.3-lacZ knock-in mice bred on the hsf1deficient genetic background (hsf1-/--hsp70.3+/--lacZ), to further elucidate the function of Hsf1 and its participation as a transcriptional activator of Hsp70 synthesis under normal or heat-induced stress conditions in vitro and in vivo. The results revealed that heat-induced Hsp70 expression in mouse tissue is entirely controlled by Hsf1, whereas its activity is not required for tissue-specific constitutive Hsp70 expression. We further demonstrate that Hsf1 is critical for maintaining cellular integrity after heat stress and that cells from hsf1-/- mice lack the ability to develop thermotolerance. This deficiency is explained by the elimination of stress-inducible Hsp70 and Hsp25 response in the absence of Hsf1 activity, leading to a lack of Hsp-mediated inhibition of apoptotic cell death via both caspase-dependent and caspase-independent pathways. The pivotal role of the Hsf1 transactivator in regulating rapid synthesis of Hsps as a critical cellular defense mechanism against environmental stress-induced damage is underlined.
- Published
- 2002
36. Critical role for perforin-, Fas/FasL-, and TNFR1-mediated cytotoxic pathways in down-regulation of antigen-specific T cells during persistent viral infection
- Author
-
Rong Ou, Demetrius Moskophidis, Shenghua Zhou, and Lei Huang
- Subjects
Cytotoxicity, Immunologic ,Pore Forming Cytotoxic Proteins ,Fas Ligand Protein ,Immunology ,bcl-X Protein ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Lymphocyte Activation ,Virus Replication ,Microbiology ,Epitope ,Fas ligand ,Receptors, Tumor Necrosis Factor ,Interleukin 21 ,Mice ,Antigen ,Antigens, CD ,Virology ,Proto-Oncogene Proteins ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,fas Receptor ,bcl-2-Associated X Protein ,Membrane Glycoproteins ,biology ,Perforin ,Mice, Inbred C57BL ,Kinetics ,Proto-Oncogene Proteins c-bcl-2 ,Receptors, Tumor Necrosis Factor, Type I ,Insect Science ,Acute Disease ,Chronic Disease ,biology.protein ,Pathogenesis and Immunity ,Tumor necrosis factor alpha ,CD8 ,Spleen ,Signal Transduction - Abstract
Viral persistence following infection with invasive strains of lymphocytic choriomeningitis virus (LCMV) can be achieved by selective down-regulation of virus-specific T lymphocytes. High viral burden in the onset of infection drives responding cells into functional unresponsiveness (anergy) that can be followed by their physical elimination. In this report, we studied down-regulation of the virus-specific CD8+-T-cell response during persistent infection of adult mice with LCMV, with emphasis on the role of perforin-, Fas/FasL-, or tumor necrosis factor receptor 1 (TNFR1)-mediated cytolysis in regulating T-cell homeostasis. The results reveal that the absence of perforin, Fas-ligand, or TNFR1 has no significant effect on the kinetics of proliferation and functional inactivation of virus-specific CD8+T cells in the onset of chronic LCMV infection. However, these molecules play a critical role in the homeostatic regulation of T cells, influencing the longevity of the virus-specific CD8+-T-cell population once it has become anergic. Thus, CD8+T cells specific to the dominant LCMV NP396–404epitope persist in an anergic state for at least 70 days in perforin-, FasL-, or TNFR1-deficient mice, but they were eliminated by day 30 in C57BL/6 controls. These effects were additive as shown by a deficit of apoptotic death of NP396–404peptide-specific CD8+T cells in mice lacking both perforin and TNFR1. This suggests a role for perforin-, FasL-, and TNFR1-mediated pathways in down-regulation of the antiviral T cell response during persistent viral infection by determining the fate of antigen-specific T cells. Moreover, virus-specific anergic CD8+T cells in persistently infected C57BL/6 mice contain higher levels of Bcl-2 and Bcl-XL than functionally intact T cells generated during acute LCMV infection. In the case of proapoptotic factors, Bax expression did not differ between T-cell populations and Bad was below the limit of detection in all samples. As expression of the Bcl-2 family members controls susceptibility to apoptosis, this finding may provide a molecular basis for the survival of anergic cells under conditions of prolonged antigen stimulation.
- Published
- 2001
37. Targeted inactivation of the sodium-calcium exchanger (Ncx1) results in the lack of a heartbeat and abnormal myofibrillar organization
- Author
-
Srinagesh V. Koushik, Rhonda Rogers, Demetrius Moskophidis, Tony L. Creazzo, Nevin A. Lambert, Jian Wang, and Simon J. Conway
- Subjects
Heartbeat ,Mice, Transgenic ,Biochemistry ,Sodium-Calcium Exchanger ,Embryonic and Fetal Development ,Mice ,Genes, Reporter ,Genetics ,Morphogenesis ,Animals ,Molecular Biology ,Gene knockdown ,Sodium-calcium exchanger ,Heart development ,Chemistry ,Myocardium ,Gene targeting ,Embryo ,Heart ,Embryonic stem cell ,Immunohistochemistry ,Myocardial Contraction ,Cell biology ,embryonic structures ,Gene Targeting ,cardiovascular system ,Calcium ,Myofibril ,Biotechnology - Abstract
SPECIFIC AIMSBecause of the following—controversial role, early heart-specific expression patterns, lack of specific inhibitors, contradictory results obtained via antisense knockdown experiments, and differing functions assigned to Na+/Ca2+ exchanger (Ncx1) during embryonic excitation-contraction coupling—we used gene targeting to delete Ncx1, which allowed us to begin to determine the precise role of Na+/Ca2+ exchange in development of the mammalian heart.PRINCIPAL FINDINGS1. Ncx1-null embryos lack a spontaneously beating heart and are embryonically lethalHeterozygous mice are grossly normal and fertile, whereas Ncx1-null embryos do not survive past ∼11.5 dpc. Ncx1-null 9.0 dpc embryos were of normal size and had undergone looping of the heart tube (Fig. 1a⤻ , b⤻ ). However, 10.0 dpc Ncx1-nulls were severely retarded in size, although they were still developing despite the lack of a heartbeat (Fig. 1c⤻ ), indicating that diffusion of growth factors/nutrients is not sufficient to maintain normal growth o...
- Published
- 2001
38. Role of virus and host variables in virus persistence or immunopathological disease caused by a non-cytolytic virus
- Author
-
Rolf M. Zinkernagel, M. van den Broek, Manuel Battegay, U. Hoffmann-Rohrer, Demetrius Moskophidis, E. Laine, University of Zurich, and Moskophidis, D
- Subjects
viruses ,T cell ,Molecular Sequence Data ,610 Medicine & health ,chemical and pharmacologic phenomena ,Biology ,Lymphocytic Choriomeningitis ,10263 Institute of Experimental Immunology ,Lymphocytic choriomeningitis ,Antibodies, Viral ,Virus ,Epitope ,Epitopes ,Mice ,Antigen ,Virology ,medicine ,Cytotoxic T cell ,Animals ,Amino Acid Sequence ,Base Sequence ,H-2 Antigens ,hemic and immune systems ,medicine.disease ,Mice, Inbred C57BL ,CTL ,medicine.anatomical_structure ,Haplotypes ,Immunology ,2406 Virology ,570 Life sciences ,biology ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
C57BL/6 mice infected with increasing doses of the Armstrong isolate of lymphocytic choriomeningitis virus (LCMV) or a variant Cl 13-Armstrong, derived from LCMV-Armstrong, exhibited distinct phenotypes with respect to clearance of virus and to cytotoxic CD8+ T cell (CTL)-dependent immunopathological disease. Low (10(2) p.f.u.) and high doses (10(7) p.f.u.) of LCMV-Armstrong were cleared rapidly from immunocompetent mice. Inoculation of a high dose (10(7) p.f.u.) of LCMV Cl 13-Armstrong temporarily induced a partial deletion of the antiviral CTL precursors (CTL-p) leading to chronic infection in several organs. Although virus was cleared from most organs by day 90-150 post-infection, it persisted in the kidney. The few remaining CTL-p were able to expand and eventually clear the virus. Systemic viral titres correlated inversely with the number of CTL-p. However, in contrast LCMV-Docile injected at high dose was able to cause complete exhaustion of CTL-p resulting in long term viral persistence. LCMV-Aggressive, derived from the same parental LCMV-WE (UBC) as Docile, showed a phenotype comparable to wild-type virus. Doses of10(7) p.f.u. of both Armstrong virus and of Cl 13-Armstrong failed to exhaust CTL-p and caused lethal CD8+ T cell-dependent choriomeningitis and a substantial footpad swelling after local infection. By contrast, doses10(3) p.f.u. of LCMV-Docile failed to cause lethal choriomeningitis in C57BL/6 mice. When Cl 13-Armstrong virus (but not LCMV-Armstrong) was injected intravenously in addition to intracerebrally or into the foot, the local immunopathology was abrogated in a dose-dependent fashion. The suppression of immunopathology paralleled the extent of exhaustion of the specific CD8+ T cell response. Nucleotide sequence analysis of the viral S-RNA fragments coding for CTL epitopes in H-2b mice revealed an asparagine to serine change of amino acid 280 in the CTL epitope 275-286 of the LCMV-Docile glycoprotein (GP) in comparison to LCMV-Aggressive or wild-type WE. This change reduced overall CTL activity and thereby probably contributes to exhaustion of CTL responses in C57BL/6 (H-2b) mice. Thus, local versus systemic antigen distribution, viral characteristics and immunological parameters determine induction and exhaustion of CD8+ T cells and the course and extent of immunopathological disease.
- Published
- 1995
39. Functional commitment to helper T cell lineage precedes positive selection and is independent of T cell receptor MHC specificity
- Author
-
Clio Mamalaki, Eugenia Spanopoulou, Dimitris Kioussis, Deborah Lans, David Baltimore, Andrea Itano, Ellen Robeyj, Demetrius Moskophidis, Mauro Tolaini, and Paola Corbella
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,T cell ,Immunology ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,Genes, MHC Class I ,Mice, Transgenic ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Interleukin 21 ,Mice ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,IL-2 receptor ,Antigen-presenting cell ,DNA Primers ,Homeodomain Proteins ,B-Lymphocytes ,Base Sequence ,ZAP70 ,CD28 ,Proteins ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Molecular biology ,Mice, Inbred C57BL ,Infectious Diseases ,medicine.anatomical_structure ,Interleukin-4 ,Peptides ,CD8 - Abstract
Thymocyte differentiation proceeds from double positive CD4+CD8+ to single positive T cells. It has been proposed that this process occurs by an instructive or a stochastic mechanism. In this report, we show that in recombination-deficient mice (RAG-1-I-) constitutive expression of a CD8 transgene allows maturation of CD4+(CD8tg+) cells, which express mature levels of a transgenic class I-restricted T cell receptor, F5. Rescued F5+CD4+(CD8tg+) cells have equivalent levels of T cell receptor expression as CD8end+ cells, respond to cognate antigen and, upon stimulation, they exhibit a phenotype characteristic of CD4+ helper T cells. These data are consistent with a model of differentiation that predicts that thymocytes become functionally committed to a helper or cytotoxic lineage before the final step of positive selection and independently of MHC specificity of their T cell receptor.
- Published
- 1994
40. Enhanced establishment of a virus carrier state in adult CD4+ T-cell-deficient mice
- Author
-
A Rahemtulla, R M Zinkernagel, Tak W. Mak, Demetrius Moskophidis, Hans Hengartner, and Manuel Battegay
- Subjects
CD4-Positive T-Lymphocytes ,Cellular immunity ,viruses ,Immunology ,chemical and pharmacologic phenomena ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis ,Antibodies, Viral ,Microbiology ,Virus ,Mice ,Immune system ,Neutralization Tests ,Virology ,medicine ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,Arenavirus ,biology ,medicine.disease ,biology.organism_classification ,Mice, Inbred C57BL ,CTL ,Insect Science ,Carrier State ,Mutation ,CD8 ,T-Lymphocytes, Cytotoxic ,Research Article - Abstract
CD4+ T cells play an important role in regulating the immune response; their contribution to virus clearance is variable. Mice that lack CD4+ T cells (CD4-/- mice) and are therefore unable to produce neutralizing antibodies cleared viscero-lymphotropic lymphocytic choriomeningitis virus (LCMV) strain WE when infected intravenously with a low dose (2 x 10(2) PFU) because of an effective CD8+ cytotoxic T-cell (CTL) response. In contrast, infection with a high dose (2 x 10(6) PFU) of LCMV strain WE led to expansion of antiviral CTL, which disappeared in CD4-/- mice; in contrast, CD4+ T-cell-competent mice developed antiviral memory CTL. This exhaustion of specific CTL caused viral persistence in CD4-/- mice, whereas CD4+ T-cell-competent mice eliminated the virus. After infection of CD4-/- mice with the faster-replicating LCMV strain DOCILE, abrogation of CTL response and establishment of viral persistence developed after infection with a low dose (5 x 10(2) PFU), i.e., an about 100-fold lower dose than in CD(4+)-competent control mice. These results show that absence of T help enhances establishment of an LCMV carrier state in selected situations.
- Published
- 1994
41. Viruses Escaping Immunological Surveillance
- Author
-
R M Zinkernagel, I Ciernik, Manuel Battegay, Demetrius Moskophidis, Diego Kyburz, Hans Hengartner, Hanspeter Pircher, Ch. Schalcher, Bernhard Odermatt, and Marie-Anne Bründler
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Rabies virus ,Rubella virus ,Biology ,medicine.disease_cause ,biology.organism_classification ,Virus ,Immunity ,Vesicular stomatitis virus ,Immunology ,medicine ,biology.protein ,Tumor necrosis factor alpha ,Antibody - Abstract
Patterns of anti-viral immunity varies from virus to virus (Mims, 1982); for recovery from some virus infections antibodies are all important (e.g. rabies virus) whereas for others T cell immunity is crucial. Protection against reinfection is mediated mostly by antibody dependent mechanisms. Of course, macrophages, interleukins (particularly IFN c, TNF etc.) possibly ADCC, NK activities etc. may play some role in both recovery from primary infection or in protection against rechallenge, but their respective roles are far less clear. These immunological parameters on one hand and the viro-logical and parasitic ones on the other hand will crucialy determine the overall outcome of an infection and of disease (summarized in Table 1).
- Published
- 1993
42. Immunosuppression by lymphocytic choriomeningitis virus infection: competent effector T and B cells but impaired antigen presentation
- Author
-
U Hoffman-Rohrer, Demetrius Moskophidis, Thomas M. Kündig, Hans Hengartner, R M Zinkernagel, Alana Althage, and Bernhard Odermatt
- Subjects
Adoptive cell transfer ,viruses ,T-Lymphocytes ,Immunology ,Antigen presentation ,Molecular Sequence Data ,Antigen-Presenting Cells ,chemical and pharmacologic phenomena ,Vaccinia virus ,Biology ,Lymphocytic Choriomeningitis ,Antibodies, Viral ,Interleukin 21 ,Mice ,T-Lymphocyte Subsets ,medicine ,Immune Tolerance ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,Antigen-presenting cell ,B cell ,B-Lymphocytes ,Base Sequence ,virus diseases ,T lymphocyte ,Virology ,medicine.anatomical_structure ,Immunoglobulin G ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
Lymphocytic choriomeningitis virus (LCMV) may cause a severe immunosuppression in mice. Its pathogenesis is apparently dependent on LCMV-specific CD8 effector T cells that mediate the destruction of virus-infected cells which are normally essentially involved in immune responses. Evaluation of various LCMV isolates in this study established a general correlation between their tropism for lymphohemopoietic cells and immunosuppression. When immune responses were assessed as the capacity of mice to mount an anti-vaccinia virus cytotoxic T cell response or an IgG response to vesicular stomatitis virus (VSV), after a primary LCMV infection, LCMV-Armstrong, WE, Clone 13 and Docile were increasingly immunosuppressive in a dose-dependent fashion with respect to both extent and duration. Analysis of lymphocyte subpopulations showed variable effects of the various LCMV isolates that did not reveal patterns readily explaining immunosuppression. To evaluate whether LCMV infection affected T and/or B cell functions directly or whether antigen presentation was impaired, adoptive transfer experiments were performed. Untreated or irradiated but uninfected normal recipient mice receiving adoptively transferred T or B cells from LCMV-WE or Docile-infected immunosuppressed donor mice responded within 30%-100% of normal ranges in both assay systems. In contrast, when T or B cells from normal donors were transferred to irradiated or non-irradiated LCMV-immunosuppressed recipients, they failed to mount a significant cytotoxic T cell response against vaccinia virus or an IgG response to VSV. Thus, the T and B cells from LCMV-immunosuppressed mice were able to function within normal ranges; in contrast, histologically and functionally, antigen presentation was severely impaired in LCMV-immunosuppressed mice.
- Published
- 1992
43. Suppression of virus-specific antibody production by CD8+ class I-restricted antiviral cytotoxic T cells in vivo
- Author
-
Hanspeter Pircher, Demetrius Moskophidis, Hans Hengartner, I Ciernik, R M Zinkernagel, and Bernhard Odermatt
- Subjects
Adoptive cell transfer ,viruses ,CD8 Antigens ,Immunology ,Antigen presentation ,Genes, MHC Class I ,chemical and pharmacologic phenomena ,Antibodies, Viral ,Microbiology ,Sensitivity and Specificity ,Animals, Genetically Modified ,Major Histocompatibility Complex ,Mice ,Viral Envelope Proteins ,Virology ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,Immunosuppression Therapy ,biology ,virus diseases ,Antiviral antibody ,hemic and immune systems ,Molecular biology ,Immunohistochemistry ,Peptide Fragments ,CTL ,Immunoglobulin M ,Insect Science ,Tissue Transplantation ,biology.protein ,Antibody ,CD8 ,Spleen ,T-Lymphocytes, Cytotoxic ,Research Article - Abstract
The question of whether virus-induced immunosuppression includes the antibody response against the infecting virus itself was evaluated in a model situation. Transgenic mice expressing the T-cell receptor (TCR) specific for peptide 32-42 of lymphocytic choriomeningitis virus (LCMV) glycoprotein 1 presented by Db reacted with a strong transgenic cytotoxic T-lymphocyte (CTL) response starting on day 3 after infection with a high dose (10(6) PFU intravenously [i.v.]) of the WE strain of LCMV (LCMV-WE); LCMV-specific antibody production in the spleen was suppressed in these mice. Low-dose (10(2) PFU i.v.) infection resulted in an antiviral antibody response comparable to that of the transgene-negative littermates. The induction of suppression of LCMV-specific antibody responses was specifically mediated by CD8+ TCR transgenic CTLs, since the LCMV-8.7 variant virus (which is not recognized by transgenic TCR-expressing CTLs because of a point mutation) did not induce suppression. In addition, treatment with CD8 monoclonal antibody in vivo abrogated suppression. Once suppression had been established, it was found to be nonspecific. The abrogation of antibody responses depended on the relative kinetics of the antibody response involved and the kinetics of the anti-LCMV CTL response. Analysis of T- and B-cell subpopulations showed no significant changes, but immunohistochemical analysis of spleens revealed extensive destruction of follicular organization in lymphoid tissue by day 4 in transgenic mice infected with LCMV-WE but not in those infected with the CTL escape mutant LCMV-8.7. Impairment of antigen presentation rather than of T or B cells was also suggested by adoptive transfer experiments, showing that transferred infected macrophages may improve the anti-LCMV antibody response in LCMV-immunosuppressed transgenic recipients; also, T and B cells from suppressed transgenic mice did respond in irradiated and virus-infected nontransgenic mice with antibody formation to LCMV. Such virus-triggered, T-cell-mediated immunopathology causing the suppression of B cells and of protective antibody responses, including those against the infecting virus itself, may permit certain viruses to establish persistent infections.
- Published
- 1992
44. In vitro selection of lymphocytic choriomeningitis virus escape mutants by cytotoxic T lymphocytes
- Author
-
Demetrius Moskophidis, Hans Hengartner, Toni Aebischer, Urs Hoffmann Rohrer, and Rolf M. Zinkernagel
- Subjects
Cytotoxicity, Immunologic ,Macromolecular Substances ,viruses ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Simian virus 40 ,Lymphocytic choriomeningitis ,Major histocompatibility complex ,Epitope ,Cell Line ,Epitopes ,Mice ,Multiplicity of infection ,medicine ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,Amino Acid Sequence ,Cell Line, Transformed ,Multidisciplinary ,biology ,Base Sequence ,T-cell receptor ,Genetic Variation ,hemic and immune systems ,T lymphocyte ,medicine.disease ,Virology ,Clone Cells ,Mice, Inbred C57BL ,CTL ,Oligodeoxyribonucleotides ,DNA, Viral ,biology.protein ,RNA, Viral ,T-Lymphocytes, Cytotoxic ,Research Article - Abstract
Cytotoxic T lymphocyte (CTL)-mediated cytolysis is induced via the interaction of the specific T-cell antigen receptor and the peptidic viral antigen associated with the major histocompatibility complex class I antigen. Here we demonstrate in vitro that lymphocytic choriomeningitis virus (LCMV) can escape the cytotoxic activity of LCMV-specific cloned CTLs by single amino acid changes within the recognized T-cell epitope defined by residues 275-289 of the LCMV glycoprotein [LCMV-GP-(275-289)]. LCMV-infected fibroblasts at a multiplicity of infection of 10(-3) exposed to virus-specific CTL at an effector-to-target cell ratio of 4:1 4 hr after infection was optimal for virus mutant selection. The selections were carried out with three LCMV-GP-(275-289)-specific CTL clones expressing T-cell antigen receptors containing the identical variable gene segments V alpha 4 and V beta 10 but different junctional regions; selection was also possible with LCMV-GP-(275-289)-specific cytotoxic polyclonal T cells. The most common escape mutation was an amino acid change of asparagine (AAT) to aspartic acid (GAT) at position 280; an additional mutation was glycine (GGT) to aspartic acid (GAT) at position 282. The results presented show that relevant point mutations within the T-cell epitope of LCMV-GP-(275-289) occur frequently and that they are selectable in vitro by CTLs.
- Published
- 1991
45. Production of random classes of immunoglobulins in brain tissue during persistent viral infection paralleled by secretion of interleukin-6 (IL-6) but not IL-4, IL-5, and gamma interferon
- Author
-
Demetrius Moskophidis, J Löhler, Adriano Fontana, Karl Frei, and R M Zinkernagel
- Subjects
medicine.medical_treatment ,Immunology ,Immunoglobulins ,Mice, Inbred Strains ,Biology ,Lymphocytic Choriomeningitis ,Antibodies, Viral ,Microbiology ,Interferon-gamma ,Mice ,Mice, Inbred AKR ,Species Specificity ,Interferon ,Virology ,medicine ,Animals ,Interferon gamma ,Interleukin 5 ,Interleukin 4 ,Interleukin-6 ,Brain ,Isotype ,Mice, Inbred C57BL ,Cytokine ,Phenotype ,Mice, Inbred DBA ,Insect Science ,Antibody Formation ,biology.protein ,Mice, Inbred CBA ,Interleukin-4 ,Antibody ,Interleukin-5 ,CD8 ,medicine.drug ,Research Article - Abstract
The activities of cytokines were determined in cerebrospinal fluid (CSF) and serum of mice persistently or intracerebrally acutely infected with lymphocytic choriomeningitis (LCM) virus (LCMV). In contrast to CBA/J (LCMV carrier) mice that responded with low levels of LCMV-specific antibody, high-responder NMRI (carrier) mice showed antibody production by B cells outside of lymphoid organs. The B cells that had infiltrated the brains of LCMV carrier mice exhibited no preferential immunoglobulin isotype or subtype virus-specific antibody production. Phenotypic analysis of the brain infiltrates in virus carrier mice revealed dominance of CD4+ T cells in contrast to virtual absence of CD4+ and dominance of CD8+ in mice with acute LCM. In NMRI but not in CBA/J carrier mice, significant concentrations of interleukin-6 (IL-6) were detected in CSF and serum; IL-2, IL-4, IL-5, granulocyte-macrophage CSF (GM-CSF), and gamma interferon (IFN-gamma) were not elevated. In contrast, during acute, lethal LCM, IL-6 and IFN-gamma were found at high concentrations, and IL-4, IL-5, and GM-CSF were detectable in CSF and serum, but virus-specific antibody-producing cells were not (yet) detectable in the brain. Thus, distinct cytokine patterns are found in acute versus chronic LCMV infection of the brain: in LCM carrier mice, local random-class immunoglobulin production correlated with the absence of IL-2, IL-4, IL-5, and IFN-gamma but active secretion of IL-6.
- Published
- 1991
46. Viral escape by selection of cytotoxic T cell-resistant virus variants in vivo
- Author
-
Hanspeter Pircher, Kurt Bürki, Urs Hoffmann Rohrer, Demetrius Moskophidis, Hans Hengartner, and Rolf M. Zinkernagel
- Subjects
viruses ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Lymphocytic Choriomeningitis ,Polymerase Chain Reaction ,Virus ,Epitope ,Epitopes ,Mice ,Immune system ,Antigen ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,Amino Acid Sequence ,Cloning, Molecular ,Neutralizing antibody ,Antigens, Viral ,Glycoproteins ,Multidisciplinary ,Arenavirus ,biology ,Base Sequence ,Genetic Variation ,biology.organism_classification ,Virology ,Immunosurveillance ,Mice, Inbred C57BL ,Immunology ,Mutation ,biology.protein ,Spleen ,T-Lymphocytes, Cytotoxic - Abstract
Viruses persist in an immune population, as in the case of influenza, or in an individual, as postulated for human immunodeficiency virus, when they are able to escape existent neutralizing antibody responses by changing their antigens. It is now shown that viruses can in principle escape the immunosurveillance of virus-specific cytotoxic T cells by mutations that alter the relevant T-cell epitope.
- Published
- 1990
47. Essential requirement for both hsf1 and hsf2 transcriptional activity in spermatogenesis and male fertility.
- Author
-
Guanghu Wang, Zhekang Ying, Xiongjie Jin, Naxin Tu, Yan Zhang, Michele Phillips, and Demetrius Moskophidis
- Published
- 2004
- Full Text
- View/download PDF
48. Targeted disruption of the heat shock transcription factor (hsf)-2 gene results in increased embryonic lethality, neuronal defects, and reduced spermatogenesis.
- Author
-
Guanghu Wang, Jing Zhang, and Demetrius Moskophidis
- Published
- 2003
- Full Text
- View/download PDF
49. The immune response of the mouse to lymphocytic choriomeningitis virus
- Author
-
Demetrius Moskophidis and Fritz Lehmann-Grube
- Subjects
biology ,Immunology ,Major histocompatibility complex ,Lymphocytic choriomeningitis ,medicine.disease ,Virology ,Virus ,CTL ,Immune system ,Inbred strain ,biology.protein ,medicine ,Cytotoxic T cell ,Cytotoxicity - Abstract
By use of an accurate procedure for the measurement of relative numbers of murine cytotoxic T lymphocytes (CTL) marked differences of lymphocytic choriomeningitis virus-specific CTL in spleens were found among mouse strains that ranged from low over intermediate and high to very high, suggesting a gradient of responses rather than distinct classes. Differences were also found between mice sharing the major histocompatibility complex.
- Published
- 1983
50. Mechanism of Recovery from Acute Virus Infection. IX. Clearance of Lymphocytic Choriomeningitis (LCM) Virus from the Feet of Mice Undergoing LCM Virus-specific Delayed-type Hypersensitivity Reaction
- Author
-
Fritz Lehmann-Grube, Lubin Fang, Demetrius Moskophidis, and Jan Gossmann
- Subjects
CD4-Positive T-Lymphocytes ,Adoptive cell transfer ,T-Lymphocytes ,chemical and pharmacologic phenomena ,Lymphocytic Choriomeningitis ,Biology ,Lymphocytic choriomeningitis ,T-Lymphocytes, Regulatory ,Virus ,Mice ,Virology ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Cytotoxic T cell ,Hypersensitivity, Delayed ,Immunity, Cellular ,Arenavirus ,Histocompatibility Antigens Class II ,Immunization, Passive ,medicine.disease ,biology.organism_classification ,Specific Pathogen-Free Organisms ,CTL ,Delayed hypersensitivity ,Immunology ,Interleukin-2 ,Female ,CD8 - Abstract
Summary As shown previously, after inoculation into the footpad of a mouse the lymphocytic choriomeningitis (LMC) virus multiplies locally. Beginning on day 6 or 7 after infection, the foot undergoes a delayed-type hypersensitivity (DTH) reaction which consists of two distinct phases that are mediated by CD8+ cells and CD4+ cells, respectively, and at about the same time the virus is eliminated. In general, for terminating infection of the mouse with LCM virus the CD8+ cytotoxic/suppressive T lymphocyte (CTL) is essential; we have now determined the cells that mediate control of the virus in a tissue undergoing a specific DTH reaction. Depletion, in infected mice, of all T lymphocytes by treatment with anti-Thy-1 monoclonal antibody prevented virus elimination from the foot, and the same was true when the CD8+ CTLs were removed. Depletion of the CD4+ helper/suppressor subset only marginally impaired the ability of the mice to rid themselves of the virus. The conclusion that here too the principal antiviral element is the CD8+ CTL was confirmed by experiments in which footpad-infected mice were adoptively immunized with virus-immune splenocytes from syngeneic mice selected for subclasses of T lymphocytes, or from mice differing in defined regions of the major histocompatibility complex (MHC), and also by experiments in which monocytes were virtually absent. However, CD8+ CTL alone or cells from MHC recombinant mice with identity in class I loci were never as antivirally active as unseparated splenocytes from syngeneic donor mice. Since the CD8+ cells' performance could be optimized by interleukin-2, we assume that the CD4+ T lymphocytes function as accessory cells; the same probably applies to monocytes.
- Published
- 1989
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.