Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.018
Filter
1.
J Transl Med ; 22(1): 63, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38229084

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Microglia-mediated neuroinflammation has been largely considered one of main factors to the PD pathology. MicroRNA-218-5p (miR-218-5p) is a microRNA that plays a role in neurodevelopment and function, while its potential function in PD and neuroinflammation remains unclear. METHODS: We explore the involvement of miR-218-5p in the PD in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model. The miR-218-5p agomir used for overexpression was delivered into the substantia nigra (SN) by bilateral stereotaxic infusions. The loss of dopaminergic (DA) neurons and microglial inflammation in the SN was determined using Western blotting and immunofluorescence. Motor function was assessed using the rotarod test. RNA sequencing (RNA-seq) was performed to explore the pathways regulated by miR-218-5p. The target genes of miR-218-5p were predicted using TargetScan and confirmed using dual luciferase reporter assays. The effects of miR-218-5p on microglial inflammation and related pathways were verified in murine microglia-like BV2 cells. To stimulate BV2 cells, SH-SY5Y cells were treated with 1-methyl-4-phenylpyridinium (MPP+) and the conditioned media (CM) were collected. RESULTS: MiR-218-5p expression was reduced in both the SN of MPTP-induced mice and MPP+-treated BV2 cells. MiR-218-5p overexpression significantly alleviated MPTP-induced microglial inflammation, loss of DA neurons, and motor dysfunction. RNA sequence and gene set enrichment analysis showed that type I interferon (IFN-I) pathways were upregulated in MPTP-induced mice, while this upregulation was reversed by miR-218-5p overexpression. A luciferase reporter assay verified that Ddx41 was a target gene of miR-218-5p. In vitro, miR-218-5p overexpression or Ddx41 knockdown inhibited the IFN-I response and expression of inflammatory cytokines in BV2 cells stimulated with MPP+-CM. CONCLUSIONS: MiR-218-5p suppresses microglia-mediated neuroinflammation and preserves DA neurons via Ddx41/IFN-I. Hence, miR-218-5p-Ddx41 is a promising therapeutic target for PD.


Subject(s)
Interferon Type I , MicroRNAs , Neuroblastoma , Parkinson Disease , Humans , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Microglia/metabolism , Neuroinflammatory Diseases , Interferon Type I/adverse effects , Interferon Type I/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Dopaminergic Neurons/metabolism , Inflammation/pathology , Dopamine/adverse effects , Dopamine/metabolism , Luciferases/metabolism , Mice, Inbred C57BL
2.
Kidney Int ; 101(6): 1142-1159, 2022 06.
Article in English | MEDLINE | ID: mdl-35339535

ABSTRACT

Type I interferon (IFN-I) mediates tissue damage in a wide range of kidney disorders, directly affecting the biology and function of several renal cell types including podocytes, mesangial, endothelial, and parietal epithelial cells. Enhanced IFN-I signaling is observed in the context of viral infections, autoimmunity (e.g., systemic lupus erythematosus), and type 1 interferonopathies, rare monogenic disorders characterized by constitutive activation of the IFN-I pathway. All these IFN-I-related disorders can cause renal dysfunction and share pathogenic and histopathological features. Collapsing glomerulopathy, a histopathological lesion characterized by podocyte loss, collapse of the vascular tuft, and parietal epithelial cell proliferation, is commonly associated with viral infections, has been described in type 1 interferonopathies such as Aicardi-Goutières syndrome and stimulator of IFN genes-associated vasculopathy with onset in infancy, and can also be induced by recombinant IFN therapy. In all these conditions, podocytes and parietal epithelial cells seem to be the primary target of IFN-I-mediated damage. Additionally, immune-mediated glomerular injury is common to viral infections, systemic lupus erythematosus, and type 1 interferonopathies such as coatomer subunit-α syndrome (COPA) and DNASE1L3 deficiency, diseases in which IFN-I apparently promotes immune-mediated kidney injury. Finally, kidney pathology primarily characterized by vascular lesions (e.g., thrombotic microangiopathy and vasculitis) is a hallmark of type 1 interferonopathy adenosine deaminase 2 deficiency as well as of systemic lupus erythematosus, viral infections, and IFN therapy. Defining the nosology, pathogenic mechanisms, and histopathological patterns of IFN-I-related kidney disorders has diagnostic and therapeutic implications, especially considering the likely near-term availability of novel drugs targeting the IFN-I pathway.


Subject(s)
Interferon Type I , Kidney Diseases , Lupus Erythematosus, Systemic , Antiviral Agents , Humans , Interferon Type I/adverse effects , Interferon Type I/metabolism , Kidney/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/genetics , Kidney Glomerulus/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics
3.
Ann Rheum Dis ; 81(7): 951-961, 2022 07.
Article in English | MEDLINE | ID: mdl-35338035

ABSTRACT

OBJECTIVES: To characterise the efficacy and safety of anifrolumab in patients with systemic lupus erythematosus (SLE) according to interferon gene signature (IFNGS), demographic and clinical subgroups. METHODS: We performed post hoc analyses of pooled data from the 52-week phase III TULIP-1/TULIP-2 placebo-controlled trials of intravenous anifrolumab in moderate-to-severe SLE. Outcomes were assessed in predefined subgroups: IFNGS (high/low), age, sex, body mass index, race, geographic region, age of onset, glucocorticoid use, disease activity and serological markers. RESULTS: In pooled data, patients received anifrolumab 300 mg (360/726) or placebo (366/726); 82.6% were IFNGS-high. IFNGS-high patients had greater baseline disease activity and were more likely to have abnormal serological markers versus IFNGS-low patients. In the total population, a greater proportion of patients treated with anifrolumab versus placebo achieved British Isles Lupus Assessment Group-based Composite Lupus Assessment (BICLA) response at week 52 (difference 16.6%; nominal p<0.001). BICLA response treatment differences with anifrolumab versus placebo were comparable to the total population across most predefined subgroups, including subgroups for baseline glucocorticoid dosage (<10/≥10 mg/day prednisone/equivalent) and for clinical disease activity (SLE Disease Activity Index 2000 score <10/≥10). Subgroups with larger treatment differences included IFNGS-high patients (18.2%), patients with abnormal baseline serological markers (23.1%) and Asian patients (29.2%). The safety profile of anifrolumab was similar across subgroups. CONCLUSIONS: Overall, this study supports the consistent efficacy and safety of anifrolumab across a range of patients with moderate-to-severe SLE. In a few subgroups, small sample sizes limited conclusions from being drawn regarding the treatment benefit with anifrolumab. TRIAL REGISTRATION NUMBER: NCT02446912, NCT02446899.


Subject(s)
Antibodies, Monoclonal, Humanized , Interferon Type I , Lupus Erythematosus, Systemic , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers , Double-Blind Method , Female , Glucocorticoids/therapeutic use , Humans , Interferon Type I/adverse effects , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Male , Treatment Outcome
4.
Sci Rep ; 11(1): 14789, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285256

ABSTRACT

To compare lupus pathogenesis in disparate tissues, we analyzed gene expression profiles of human discoid lupus erythematosus (DLE) and lupus nephritis (LN). We found common increases in myeloid cell-defining gene sets and decreases in genes controlling glucose and lipid metabolism in lupus-affected skin and kidney. Regression models in DLE indicated increased glycolysis was correlated with keratinocyte, endothelial, and inflammatory cell transcripts, and decreased tricarboxylic (TCA) cycle genes were correlated with the keratinocyte signature. In LN, regression models demonstrated decreased glycolysis and TCA cycle genes were correlated with increased endothelial or decreased kidney cell transcripts, respectively. Less severe glomerular LN exhibited similar alterations in metabolism and tissue cell transcripts before monocyte/myeloid cell infiltration in some patients. Additionally, changes to mitochondrial and peroxisomal transcripts were associated with specific cells rather than global signal changes. Examination of murine LN gene expression demonstrated metabolic changes were not driven by acute exposure to type I interferon and could be restored after immunosuppression. Finally, expression of HAVCR1, a tubule damage marker, was negatively correlated with the TCA cycle signature in LN models. These results indicate that altered metabolic dysfunction is a common, reversible change in lupus-affected tissues and appears to reflect damage downstream of immunologic processes.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Lupus Erythematosus, Discoid/genetics , Lupus Nephritis/genetics , Animals , Citric Acid Cycle , Databases, Genetic , Disease Models, Animal , Female , Gene Expression Regulation , Glucose/metabolism , Glycolysis , Humans , Interferon Type I/adverse effects , Lipid Metabolism , Lupus Erythematosus, Discoid/metabolism , Lupus Nephritis/metabolism , Mice
5.
Phytomedicine ; 84: 153495, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33611210

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is a chronic relapsing inflammatory disease that markedly elevates the risk of colon cancers and results in disability. The disrupted immune homeostasis has been recognized as a predominant player in the pathogenesis of UC. However, the overall remission rate of current therapies based on immunoregulation is still unsatisfactory. Si-Ni-San (SNS) has been found effective in relieving UC through thousands of years of clinical practice, yet the specific mechanisms of the protective effect of SNS were not fully elucidated. PURPOSE: We aim to investigate the therapeutic effects of SNS against the development of chronic colitis and the underlying mechanisms. METHODS: We established a DSS-induced chronic experimental colitis mouse model to evaluate the effect of SNS. RNA-sequencing, bioinformatic analysis, and in vitro studies were performed to investigate the underlying mechanisms. RESULTS: Our data demonstrated that SNS significantly ameliorated chronic experimental colitis via inhibiting the expression of genes associated with inflammatory responses. Interestingly, SNS significantly suppressed DSS-induced type I interferon (IFN) responses instead of directly downregulating the production of pro-inflammatory cytokines, such as Il-6. In vitro study further found that SNS selectively inhibited STING and RIG-I pathway-induced type I IFN responses by modulating TBK1- and IRF3-dependent signaling transduction. SNS also suppressed the expression of IFN-stimulated genes by directly inhibiting STAT1 and STAT2 activation. CONCLUSION: Our study not only provides novel insights into the pathogenic role of type I IFN responses in colitis but also suggested that SNS or bioactive compounds derived from SNS may serve as novel therapeutic strategies for the treatment of UC via interfering type I IFN-mediated inflammation.


Subject(s)
Colitis/drug therapy , Drugs, Chinese Herbal/therapeutic use , Inflammation/drug therapy , Animals , Chronic Disease , Colitis/chemically induced , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Humans , Interferon Type I/adverse effects , Mice , Neoplasm Recurrence, Local , Signal Transduction/drug effects
6.
mBio ; 11(5)2020 09 10.
Article in English | MEDLINE | ID: mdl-32913009

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), is a recently emerged respiratory coronavirus that has infected >23 million people worldwide with >800,000 deaths. Few COVID-19 therapeutics are available, and the basis for severe infections is poorly understood. Here, we investigated properties of type I (ß), II (γ), and III (λ1) interferons (IFNs), potent immune cytokines that are normally produced during infection and that upregulate IFN-stimulated gene (ISG) effectors to limit virus replication. IFNs are already in clinical trials to treat COVID-19. However, recent studies highlight the potential for IFNs to enhance expression of host angiotensin-converting enzyme 2 (ACE2), suggesting that IFN therapy or natural coinfections could exacerbate COVID-19 by upregulating this critical virus entry receptor. Using a cell line model, we found that beta interferon (IFN-ß) strongly upregulated expression of canonical antiviral ISGs, as well as ACE2 at the mRNA and cell surface protein levels. Strikingly, IFN-λ1 upregulated antiviral ISGs, but ACE2 mRNA was only marginally elevated and did not lead to detectably increased ACE2 protein at the cell surface. IFN-γ induced the weakest ISG response but clearly enhanced surface expression of ACE2. Importantly, all IFN types inhibited SARS-CoV-2 replication in a dose-dependent manner, and IFN-ß and IFN-λ1 exhibited potent antiviral activity in primary human bronchial epithelial cells. Our data imply that type-specific mechanisms or kinetics shape IFN-enhanced ACE2 transcript and cell surface levels but that the antiviral action of IFNs against SARS-CoV-2 counterbalances any proviral effects of ACE2 induction. These insights should aid in evaluating the benefits of specific IFNs, particularly IFN-λ, as repurposed therapeutics.IMPORTANCE Repurposing existing, clinically approved, antiviral drugs as COVID-19 therapeutics is a rapid way to help combat the SARS-CoV-2 pandemic. Interferons (IFNs) usually form part of the body's natural innate immune defenses against viruses, and they have been used with partial success to treat previous new viral threats, such as HIV, hepatitis C virus, and Ebola virus. Nevertheless, IFNs can have undesirable side effects, and recent reports indicate that IFNs upregulate the expression of host ACE2 (a critical entry receptor for SARS-CoV-2), raising the possibility that IFN treatments could exacerbate COVID-19. Here, we studied the antiviral- and ACE2-inducing properties of different IFN types in both a human lung cell line model and primary human bronchial epithelial cells. We observed differences between IFNs with respect to their induction of antiviral genes and abilities to enhance the cell surface expression of ACE2. Nevertheless, all the IFNs limited SARS-CoV-2 replication, suggesting that their antiviral actions can counterbalance increased ACE2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Interferons/pharmacology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Aged , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/immunology , COVID-19 , Cell Line , Chlorocebus aethiops , Female , Humans , Immunotherapy/methods , Interferon Type I/adverse effects , Interferon-gamma/adverse effects , Interferons/adverse effects , Pandemics , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/virology , SARS-CoV-2 , Up-Regulation/drug effects , Vero Cells , Virus Replication/drug effects , Interferon Lambda
7.
Int Immunopharmacol ; 86: 106740, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32645630

ABSTRACT

BACKGROUND: There is no vaccine or specific antiviral treatment for HCoVs infection. The use of type I interferons for coronavirus is still under great debate in clinical practice. MATERIALS AND METHODS: A literature search of all relevant studies published on PubMed, Cochrane library, Web of Science database, Science Direct, Wanfang Data, and China National Knowledge Infrastructure (CNKI) until February 2020 was performed. RESULTS: Of the 1081 identified articles, only 15 studies were included in the final analysis. Comorbidities and delay in diagnosis were significantly associated with case mortality. Type I interferons seem to improve respiratory distress, relieve lung abnormalities, present better saturation, reduce needs for supplemental oxygen support. Type I interferons seem to be well tolerated, and don't increase life threating adverse effects. Data on IFNs in HCoVs are limited, heterogenous and mainly observational. CONCLUSIONS: Current data do not allow making regarding robust commendations for the use of IFNs in HCoVs in general or in specific subtype. But we still recommend type I interferons serving as first-line antivirals in HCoVs infections within local protocols, and interferons may be adopted to the treatments of the SARS-CoV-2 as well. Well-designed large-scale prospective randomized control trials are greatly needed to provide more robust evidence on this topic.


Subject(s)
Antiviral Agents/administration & dosage , Betacoronavirus/immunology , Coronavirus Infections/drug therapy , Interferon Type I/administration & dosage , Pneumonia, Viral/drug therapy , Antiviral Agents/adverse effects , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/mortality , Coronavirus Infections/virology , Humans , Interferon Type I/adverse effects , Middle East Respiratory Syndrome Coronavirus , Observational Studies as Topic , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2 , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/epidemiology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/virology , Survival Analysis , Treatment Outcome , COVID-19 Drug Treatment
8.
Clin Exp Rheumatol ; 37 Suppl 118(3): 185-191, 2019.
Article in English | MEDLINE | ID: mdl-31376268

ABSTRACT

Type I interferons (IFN) have long been recognised as mediators of innate immune defense mechanisms against viral threats. Robust evidence over the last 15 years revealed their significant role in the pathogenesis of systemic autoimmune diseases, including systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS). Despite the progress, methods of detection, initial triggers, biological functions and clinical associations in the setting of autoimmunity remain to be fully clarified. As therapeutic options for SS are currently limited, neutralising specific targets of the type I IFN pathway seems a promising option. In this review we summarise the current evidence regarding the role of type I IFN in SS.


Subject(s)
Interferon Type I , Sjogren's Syndrome , Humans , Interferon Type I/adverse effects , Interferon Type I/genetics , Interferon Type I/physiology , Sjogren's Syndrome/etiology , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology
9.
Immunity ; 50(4): 907-923, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995506

ABSTRACT

Type I interferons (IFNs) (IFN-α, IFN-ß) and type III IFNs (IFN-λ) share many properties, including induction by viral infection, activation of shared signaling pathways, and transcriptional programs. However, recent discoveries have revealed context-specific functional differences. Here, we provide a comprehensive review of type I and type III IFN activities, highlighting shared and distinct features from molecular mechanisms through physiological responses. Beyond discussing canonical antiviral functions, we consider the adaptive immune priming, anti-tumor, and autoimmune functions of IFNs. We discuss a model wherein type III IFNs serve as a front-line defense that controls infection at epithelial barriers while minimizing damaging inflammatory responses, reserving the more potent type I IFN response for when local responses are insufficient. In this context, we discuss current therapeutic applications targeting these cytokine pathways and highlight gaps in understanding of the biology of type I and type III IFNs in health and disease.


Subject(s)
Interferon Type I/immunology , Interferons/immunology , Adaptive Immunity , Animals , Antiviral Agents/therapeutic use , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Epithelial Cells/immunology , Female , Humans , Interferon Type I/adverse effects , Interferon Type I/therapeutic use , Interferons/adverse effects , Interferons/therapeutic use , Male , Maternal-Fetal Exchange/immunology , Mice , Neoplasms/drug therapy , Neoplasms/immunology , Organ Specificity , Pregnancy , Signal Transduction/immunology , Transcription, Genetic , Transcriptome , Virus Diseases/drug therapy , Virus Diseases/immunology , Interferon Lambda
10.
Med Sci (Paris) ; 35(3): 232-235, 2019 Mar.
Article in French | MEDLINE | ID: mdl-30931907

ABSTRACT

In this brief review, the authors present a history of the different aspects of the scientific puzzle leading from pioneer animal studies and astute clinical experimental observations to a mature appreciation of the deleterious role of excess of a type I interferon in human pathology.


Subject(s)
Interferon Type I/adverse effects , Interferon Type I/physiology , Animals , Antiviral Agents/adverse effects , Antiviral Agents/metabolism , History, 20th Century , History, 21st Century , Humans , Immune System Diseases/etiology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/immunology , Mice , Models, Animal
12.
Blood ; 128(24): 2824-2833, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27663672

ABSTRACT

Many drugs have been reported to cause thrombotic microangiopathy (TMA), yet evidence supporting a direct association is often weak. In particular, TMA has been reported in association with recombinant type I interferon (IFN) therapies, with recent concern regarding the use of IFN in multiple sclerosis patients. However, a causal association has yet to be demonstrated. Here, we adopt a combined clinical and experimental approach to provide evidence of such an association between type I IFN and TMA. We show that the clinical phenotype of cases referred to a national center is uniformly consistent with a direct dose-dependent drug-induced TMA. We then show that dose-dependent microvascular disease is seen in a transgenic mouse model of IFN toxicity. This includes specific microvascular pathological changes seen in patient biopsies and is dependent on transcriptional activation of the IFN response through the type I interferon α/ß receptor (IFNAR). Together our clinical and experimental findings provide evidence of a causal link between type I IFN and TMA. As such, recombinant type I IFN therapies should be stopped at the earliest stage in patients who develop this complication, with implications for risk mitigation.


Subject(s)
Interferon Type I/adverse effects , Microvessels/drug effects , Thrombotic Microangiopathies/chemically induced , Animals , Biopsy , Humans , Kidney/drug effects , Kidney/pathology , Mice, Transgenic , Microvessels/ultrastructure , Multiple Sclerosis/pathology , Signal Transduction/drug effects , Species Specificity
13.
Cytokine Growth Factor Rev ; 25(4): 369-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25156421

ABSTRACT

The interferons (IFNs) are glycoproteins with strong antiviral activities that represent one of the first lines of host defense against invading pathogens. These proteins are classified into three groups, Type I, II and III IFNs, based on the structure of their receptors on the cell surface. Due to their ability to modulate immune responses, they have become attractive therapeutic options to control chronic virus infections. In combination with other drugs, Type I IFNs are considered as "standard of care" in suppressing Hepatitis C (HCV) and Hepatitis B (HBV) infections, while Type III IFN has generated encouraging results as a treatment for HCV infection in phase III clinical trials. However, though effective, using IFNs as a treatment is not without the need for caution. IFNs are such powerful cytokines that affect a wide array of cell types; as a result, patients usually experience unpleasant symptoms, with a percentage of patients suffering system wide effects. Thus, constant monitoring is required for patients treated with IFN in order to reach the treatment goals of suppressing virus infection and maintaining quality of life.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis B/drug therapy , Hepatitis C/drug therapy , Immunotherapy/methods , Interferons/therapeutic use , Antiviral Agents/adverse effects , Hepatitis B/immunology , Hepatitis C/immunology , Humans , Interferon Type I/adverse effects , Interferon Type I/therapeutic use , Interferon-gamma/adverse effects , Interferon-gamma/therapeutic use , Interferons/adverse effects , Interleukins/adverse effects , Interleukins/therapeutic use , Receptors, Interferon/classification , Signal Transduction/drug effects
14.
Qual Life Res ; 23(3): 877-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24036634

ABSTRACT

PURPOSE: To describe the psychometric properties and identify the minimally important difference (MID) of the hepatitis C virus patient-reported outcomes (HCV-PRO) instrument. Chronic HCV infection and associated treatments negatively affect PROs of function and well-being. METHODS: In a phase 2 trial, HCV-infected patients received direct-acting antivirals (DAAs) for 12 weeks with peg-interferon/ribavirin (peg-IFN/RBV) for 48 weeks, or placebo plus peg-IFN/RBV. The HCV-PRO total score, SF-36 PCS and MCS scores, EQ-5D-3L, and EQ VAS were measured at baseline, week 8, end of DAA treatment (EODT), end of peg-IFN/RBV treatment (EOT), and posttreatment week 24 (SVR24). Convergent validity of the HCV-PRO was assessed by Pearson's correlation coefficients. Discriminant validity was assessed by analyzing mean HCV-PRO total scores by EQ-5D anxiety/depression and pain/discomfort domain scores (none vs. some) and presence/absence of depression or fatigue adverse events. MID was identified through effect size (ES) and receiver-operating characteristic (ROC) curve analyses (HCV-PRO response vs. SF-36 PCS/MCS and EQ VAS MID thresholds). RESULTS: In 74 patients (22 % female; 81 % White; 51 % ≥50 years), correlations (0.64-0.96) between HCV-PRO total scores, SF-36 PCS/MCS scores, and EQ VAS scores at all time points supported convergent validity. HCV-PRO total scores were reduced to 10-30 points in patients impaired by depression, pain, or fatigue symptoms. Impact of peg-IFN/RBV regimen on HCV-PRO ES increased over time (EODT -0.76; EOT -0.93). ES and ROC curve analyses indicated an MID of -10 points. CONCLUSION: The HCV-PRO was valid and responsive in the population studied. An MID of -10 points represented a threshold of clinical significance for the HCV-PRO.


Subject(s)
Health Status Indicators , Hepatitis C, Chronic/psychology , Patient Outcome Assessment , Psychometrics/standards , Quality of Life , Adolescent , Adult , Aged , Aged, 80 and over , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Anxiety/complications , Data Interpretation, Statistical , Depression/complications , Drug Therapy, Combination , Female , Hepacivirus/classification , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Humans , Interferon Type I/adverse effects , Interferon Type I/therapeutic use , Male , Middle Aged , Placebos , Reproducibility of Results , Ribavirin/adverse effects , Ribavirin/therapeutic use , Severity of Illness Index , Surveys and Questionnaires , Treatment Outcome , Young Adult
16.
J Interferon Cytokine Res ; 33(4): 211-25, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23570388

ABSTRACT

After several decades of intense clinical research, the great promise of Type I interferons (IFN1) as the anticancer wonder drugs that could cure or, at the very least, curb the progression of various oncological diseases has regrettably failed to deliver. Severe side effects and low efficacy of IFN1-based pharmaceutics greatly limited use of these drugs and further reduced the enthusiasm of clinical oncologists for future optimization of IFN1-based therapeutic modalities. Incredibly, extensive clinical studies to assess the efficacy of IFN1 alone or in combination with other anticancer drugs have not been paralleled by an equal scope in defining the determinants that confer cell sensitivity or refractoriness to IFN1. Given that all effects of IFN1 on malignant and benign cells alike are mediated by its receptor, the mechanisms regulating these receptor cell surface levels should play a paramount role in shaping the magnitude and duration of IFN1-elicited effects. These mechanisms and their role in controlling IFN1 responses, as well as an ability of a growing tumor to commandeer these events, are the focus of our review. We postulate that activation of numerous signaling pathways leading to elimination of IFN1 receptor occurs in cancer cells and benign cells that contribute to tumor tissue. We further hypothesize that activation of these eliminative pathways enables the escape from IFN1-driven suppression of tumorigenesis and elicits the primary refractoriness of tumor to the pharmaceutical IFN1.


Subject(s)
Interferon Type I/adverse effects , Interferon Type I/therapeutic use , Neoplasms/drug therapy , Receptors, Interferon/metabolism , Drug Resistance, Neoplasm , Humans , Receptor, Interferon alpha-beta/metabolism , Signal Transduction
17.
Autoimmun Rev ; 12(5): 558-66, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23201923

ABSTRACT

Sjögren's syndrome is a chronic autoimmune disease characterized by lymphocytic infiltration of the salivary and lachrymal glands resulting in dry eyes and mouth. Genetic predisposition, pathogenic infections and hormones have been implicated in the pathogenesis of the disease. Studies in the last several years have revealed marked over-expression of the type I interferon (IFN)-inducible genes in the peripheral blood and salivary glands of patients with Sjögren's syndrome. The expression of the type I IFN-inducible genes in Sjögren's syndrome also positively correlates to titers of anti-Ro and anti-La autoantibodies, which are typical for this disease. Plasmacytoid dendritic cells (pDC) are the major source of type I IFN production and activated pDC are detected in minor salivary gland biopsies from patients with primary Sjögren's syndrome. In addition, polymorphisms in genes important both for the production and response to type I IFN are associated to increased risk for Sjögren's syndrome. Because type I IFN bears a variety of biological functions, such as defense against viral infections and activation of the immune system, these results suggest that the type I IFN system has an important role in the pathogenesis of Sjögren's syndrome. A variety of mechanisms causing an activation of the type I IFN system are discussed in this review. Given the pivotal role of type I IFN in the disease process, therapeutic interventions targeting the type I IFN signaling pathway have the potential to benefit the patients with elevated type I IFN status and such hypothesis needs to be carefully evaluated in clinical development.


Subject(s)
Interferon Type I/physiology , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology , Humans , Interferon Type I/adverse effects , Interferon Type I/genetics , Sjogren's Syndrome/etiology
18.
Intern Med ; 51(18): 2625-9, 2012.
Article in English | MEDLINE | ID: mdl-22989839

ABSTRACT

A 60-year-old woman developed type 1 diabetes mellitus and anti-aquaporin-4 antibody positive optic neuritis during type 1 interferon therapies for chronic hepatitis C. The diabetes mellitus was elicited by interferon-α plus ribavirin therapy, while the optic neuritis was induced after interferon-ß treatment, followed by interferon-α and ribavirin therapy. It is possible that type 1 interferons lead to the onset of the two autoimmune diseases by inducing disease-specific autoantibodies. Autoimmune disease is an infrequent complication of type 1 interferon treatment; however, once it has occurred, it may result in severe impairments. Patients undergoing type 1 interferon therapy should therefore be carefully monitored for any manifestations of autoimmune diseases.


Subject(s)
Antibodies, Anti-Idiotypic/blood , Aquaporin 4/immunology , Diabetes Mellitus, Type 1/chemically induced , Hepatitis C, Chronic/drug therapy , Interferon Type I/adverse effects , Interferon Type I/therapeutic use , Optic Neuritis/chemically induced , Optic Neuritis/immunology , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Diabetes Mellitus, Type 1/diagnosis , Drug Synergism , Drug Therapy, Combination , Female , Humans , Interferon-alpha/adverse effects , Interferon-alpha/therapeutic use , Interferon-beta/adverse effects , Interferon-beta/therapeutic use , Middle Aged , Optic Neuritis/diagnosis , Ribavirin/therapeutic use
19.
Immunol Rev ; 248(1): 23-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22725952

ABSTRACT

In multiple sclerosis, type I interferon (IFN) is considered immune-modulatory, and recombinant forms of IFN-ß are the most prescribed treatment for this disease. This is in contrast to most other autoimmune disorders, because type I IFN contributes to the pathologies. Even within the relapsing-remitting multiple sclerosis (RRMS) population, 30-50% of MS patients are non-responsive to this treatment, and it consistently worsens neuromyelitis optica, a disease similar to RRMS. In this article, we discuss the recent advances in the field of autoimmunity and introduce the theory explain how type I IFNs can be pro-inflammatory in disease that is predominantly driven by a Th17 response and are therapeutic when disease is predominantly Th1.


Subject(s)
Autoimmune Diseases/drug therapy , Interferon Type I/therapeutic use , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Disease Models, Animal , Encephalomyelitis/drug therapy , Encephalomyelitis/immunology , Encephalomyelitis/metabolism , Humans , Interferon Type I/adverse effects , Interferon Type I/metabolism , Interferon-beta/adverse effects , Interferon-beta/metabolism , Interferon-beta/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
20.
J Interferon Cytokine Res ; 32(3): 95-102, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22132684

ABSTRACT

This review summarizes and analyzes the clinical outcomes following treatment of a wide range of diseases with recombinant interferons (r-IFNs) and/or natural interferons (n-IFNs). The investigation focuses on the frequency of neutralizing antibodies (NABs) directed against IFN, which are formed during treatment and their clinical impact. r-IFNs (α-2a, α-2b, ß-1a, and ß-1b) induced seroconversion with generation of NABs in 17.2% of patients studied. The highest incidence of NABs occurred in macular degeneration (61.4%) with the lowest in multiple sclerosis (14.7%). The incidence of antibodies induced against n-IFNs was very low (<0.2%) and was significantly less than that seen for r-IFNs (P<0.0001). Overall, the fraction of relapsed and refractory patients is statistically greater in NAB positive patients compared to NAB negative patients (<0.0001), whereas the percentage of responding patients is higher in the NAB negative cohort (P<0.001). Finally, we also analyzed relapsed and refractory NAB positive patients who switched treatment to n-IFN, such as leukocyte derived Alferon N Injection® (α-n3) or Wellferon® (α-n1). Overall, in 33/40 (82%) of these relapsed or refractory patients, switching to n-IFNs restored the clinical response. This result is consistent with serology studies showing that the NABs directed against r-IFNs do not effectively cross-react with n-IFNs.


Subject(s)
Antibodies, Neutralizing/immunology , Interferon Type I/immunology , Cross Reactions , Humans , Incidence , Interferon Type I/adverse effects , Interferon Type I/therapeutic use , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...