Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Oncol ; 13: 1043694, 2023.
Article in English | MEDLINE | ID: mdl-37114129

ABSTRACT

Introduction: Chronic lymphocytic leukemia (CLL) is characterized by an aberrant cytokine network that can support tumor growth by triggering janus kinase (JAK)/STAT pathways. Targeting cytokine-signaling should then be a rational therapeutic strategy but the JAK inhibitor ruxolitinib failed to control and seemingly accelerated the disease in clinical trials. Methods: The effect of ruxolitinib on primary human CLL cells was studied in vitro and in vivo. Results: Ruxolitinib increased phosphorylation of IRAK4, an important toll-like receptor (TLR)- signaling intermediate, in circulating CLL cells in vitro. It also enhanced p38 and NFKB1 phosphorylation while lowering STAT3 phosphorylation in CLL cells activated with TLR-7/8 agonists and IL-2. Among the cytokines made by activated CLL cells, high levels of IL-10 contributed strongly to STAT3 phosphorylation and inhibited TLR7 activity. Ruxolitinib limited TLR-mediated IL10 transcription and markedly reduced IL-10 production in vitro. It also decreased blood levels of IL-10 while increasing TNFα along with phospho-p38 expression and gene sets associated with TLR-activation in CLL cells in vivo. The bruton's tyrosine kinase inhibitor ibrutinib decreased IL-10 production in vitro but, in contrast to ruxolitinib, blocked initial IL10 transcription induced by TLR-signaling in vitro, decreased TNFα production, and deactivates CLL cells in vivo. Discussion: These findings suggest the possible benefits of inhibiting growth factors with JAK inhibitors in CLL are outweighed by negative effects on potential tumor suppressors such as IL-10 that allow unrestrained activation of NFκB by drivers such as TLRs. Specific inhibition of growth-promoting cytokines with blocking antibodies or infusing suppressive cytokines like IL-10 might be better strategies to manipulate cytokines in CLL.

2.
Proc Natl Acad Sci U S A ; 119(20): e2117184119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35549552

ABSTRACT

Gaze understanding­a suggested precursor for understanding others' intentions­requires recovery of gaze direction from the observed person's head and eye position. This challenging computation is naturally acquired at infancy without explicit external guidance, but can it be learned later if vision is extremely poor throughout early childhood? We addressed this question by studying gaze following in Ethiopian patients with early bilateral congenital cataracts diagnosed and treated by us only at late childhood. This sight restoration provided a unique opportunity to directly address basic issues on the roles of "nature" and "nurture" in development, as it caused a selective perturbation to the natural process, eliminating some gaze-direction cues while leaving others still available. Following surgery, the patients' visual acuity typically improved substantially, allowing discrimination of pupil position in the eye. Yet, the patients failed to show eye gaze-following effects and fixated less than controls on the eyes­two spontaneous behaviors typically seen in controls. Our model for unsupervised learning of gaze direction explains how head-based gaze following can develop under severe image blur, resembling preoperative conditions. It also suggests why, despite acquiring sufficient resolution to extract eye position, automatic eye gaze following is not established after surgery due to lack of detailed early visual experience. We suggest that visual skills acquired in infancy in an unsupervised manner will be difficult or impossible to acquire when internal guidance is no longer available, even when sufficient image resolution for the task is restored. This creates fundamental barriers to spontaneous vision recovery following prolonged deprivation in early age.


Subject(s)
Fixation, Ocular , Vision, Ocular , Attention , Blindness , Child , Humans , Visual Acuity
3.
ACS Biomater Sci Eng ; 8(6): 2553-2563, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35608934

ABSTRACT

Delivering medication to the lungs via nebulization of pharmaceuticals is a noninvasive and efficient therapy route, particularly for respiratory diseases. The recent worldwide severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) pandemic urges the development of such therapies as an effective alternative to vaccines. The main difficulties in using inhalation therapy are the development of effective medicine and methods to stabilize the biological molecules and transfer them to the lungs efficiently following nebulization. We have developed a high-affinity angiotensin-converting enzyme 2 (ACE2) receptor-binding domain (RBD-62) that can be used as a medication to inhibit infection with SARS-CoV-2 and its variants. In this study, we established a nebulization protocol for drug delivery by inhalation using two commercial vibrating mesh (VM) nebulizers (Aerogen Solo and PARI eFlow) that generate similar mist size distribution in a size range that allows efficient deposition in the small respiratory airway. In a series of experiments, we show the high activity of RBD-62, interferon-α2 (IFN-α2), and other proteins following nebulization. The addition of gelatin significantly stabilizes the proteins and enhances the fractions of active proteins after nebulization, minimizing the medication dosage. Furthermore, hamster inhalation experiments verified the feasibility of the protocol in pulmonary drug delivery. In short, the gelatin-modified RBD-62 formulation in coordination with VM nebulizer can be used as a therapy to cure SARS-CoV-2.


Subject(s)
COVID-19 Drug Treatment , Gelatin , Aerosols/chemistry , Humans , Lung , SARS-CoV-2
5.
PLoS Pathog ; 17(8): e1009800, 2021 08.
Article in English | MEDLINE | ID: mdl-34437657

ABSTRACT

Type I Interferons (IFN-Is) are a family of cytokines which play a major role in inhibiting viral infection. Resultantly, many viruses have evolved mechanisms in which to evade the IFN-I response. Here we tested the impact of expression of 27 different SARS-CoV-2 genes in relation to their effect on IFN production and activity using three independent experimental methods. We identified six gene products; NSP6, ORF6, ORF7b, NSP1, NSP5 and NSP15, which strongly (>10-fold) blocked MAVS-induced (but not TRIF-induced) IFNß production. Expression of the first three of these SARS-CoV-2 genes specifically blocked MAVS-induced IFNß-promoter activity, whereas all six genes induced a collapse in IFNß mRNA levels, corresponding with suppressed IFNß protein secretion. Five of these six genes furthermore suppressed MAVS-induced activation of IFNλs, however with no effect on IFNα or IFNγ production. In sharp contrast, SARS-CoV-2 infected cells remained extremely sensitive to anti-viral activity exerted by added IFN-Is. None of the SARS-CoV-2 genes were able to block IFN-I signaling, as demonstrated by robust activation of Interferon Stimulated Genes (ISGs) by added interferon. This, despite the reduced levels of STAT1 and phospho-STAT1, was likely caused by broad translation inhibition mediated by NSP1. Finally, we found that a truncated ORF7b variant that has arisen from a mutant SARS-CoV-2 strain harboring a 382-nucleotide deletion associating with mild disease (Δ382 strain identified in Singapore & Taiwan in 2020) lost its ability to suppress type I and type III IFN production. In summary, our findings support a multi-gene process in which SARS-CoV-2 blocks IFN-production, with ORF7b as a major player, presumably facilitating evasion of host detection during early infection. However, SARS-CoV-2 fails to suppress IFN-I signaling thus providing an opportunity to exploit IFN-Is as potential therapeutic antiviral drugs.


Subject(s)
Interferon-beta/metabolism , SARS-CoV-2/immunology , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Chlorocebus aethiops , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Interferon-beta/genetics , Interferon-beta/pharmacology , SARS-CoV-2/drug effects , STAT1 Transcription Factor/metabolism , Vero Cells , Viral Proteins/genetics
6.
Nat Microbiol ; 6(9): 1188-1198, 2021 09.
Article in English | MEDLINE | ID: mdl-34400835

ABSTRACT

SARS-CoV-2 variants of interest and concern will continue to emerge for the duration of the COVID-19 pandemic. To map mutations in the receptor-binding domain (RBD) of the spike protein that affect binding to angiotensin-converting enzyme 2 (ACE2), the receptor for SARS-CoV-2, we applied in vitro evolution to affinity-mature the RBD. Multiple rounds of random mutagenic libraries of the RBD were sorted against decreasing concentrations of ACE2, resulting in the selection of higher affinity RBD binders. We found that mutations present in more transmissible viruses (S477N, E484K and N501Y) were preferentially selected in our high-throughput screen. Evolved RBD mutants include prominently the amino acid substitutions found in the RBDs of B.1.620, B.1.1.7 (Alpha), B1.351 (Beta) and P.1 (Gamma) variants. Moreover, the incidence of RBD mutations in the population as presented in the GISAID database (April 2021) is positively correlated with increased binding affinity to ACE2. Further in vitro evolution increased binding by 1,000-fold and identified mutations that may be more infectious if they evolve in the circulating viral population, for example, Q498R is epistatic to N501Y. We show that our high-affinity variant RBD-62 can be used as a drug to inhibit infection with SARS-CoV-2 and variants Alpha, Beta and Gamma in vitro. In a model of SARS-CoV-2 challenge in hamster, RBD-62 significantly reduced clinical disease when administered before or after infection. A 2.9 Å cryo-electron microscopy structure of the high-affinity complex of RBD-62 and ACE2, including all rapidly spreading mutations, provides a structural basis for future drug and vaccine development and for in silico evaluation of known antibodies.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/administration & dosage , COVID-19/virology , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Animals , Antiviral Agents/chemistry , COVID-19/genetics , COVID-19/metabolism , Cricetinae , Drug Design , Evolution, Molecular , Female , Humans , Male , Mesocricetus , Molecular Dynamics Simulation , Mutation , Protein Binding/drug effects , Protein Domains , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Virus Internalization/drug effects , COVID-19 Drug Treatment
7.
J Immunol ; 205(10): 2629-2639, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33067379

ABSTRACT

The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib has profound activity in chronic lymphocytic leukemia (CLL) but limited curative potential by itself. Residual signaling pathways that maintain survival of CLL cells might be targeted to improve ibrutinib's therapeutic activity, but the nature of these pathways is unclear. Ongoing activation of IFN receptors in patients on ibrutinib was suggested by the presence of type I and II IFN in blood together with the cycling behavior of IFN-stimulated gene (ISG) products when IFN signaling was blocked intermittently with the JAK inhibitor ruxolitinib. IFN signaling in CLL cells from human patients was not prevented by ibrutinib in vitro or in vivo, but ISG expression was significantly attenuated in vitro. ISGs such as CXCL10 that require concomitant activation of NF-κB were decreased when this pathway was inhibited by ibrutinib. Other ISGs, exemplified by LAG3, were decreased as a result of inhibited protein translation. Effects of IFN on survival remained intact as type I and II IFN-protected CLL cells from ibrutinib in vitro, which could be prevented by ruxolitinib and IFNR blocking Abs. These observations suggest that IFNs may help CLL cells persist and specific targeting of IFN signaling might deepen clinical responses of patients on ibrutinib.


Subject(s)
Adenine/analogs & derivatives , Interferon Type I/metabolism , Interferon-gamma/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines/pharmacology , Adenine/pharmacology , Adenine/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cell Survival/drug effects , Cell Survival/immunology , Drug Resistance, Neoplasm/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Nitriles , Piperidines/therapeutic use , Primary Cell Culture , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Cells, Cultured
8.
J Cogn Neurosci ; 31(9): 1354-1367, 2019 09.
Article in English | MEDLINE | ID: mdl-31059350

ABSTRACT

Visual object recognition is performed effortlessly by humans notwithstanding the fact that it requires a series of complex computations, which are, as yet, not well understood. Here, we tested a novel account of the representations used for visual recognition and their neural correlates using fMRI. The rationale is based on previous research showing that a set of representations, termed "minimal recognizable configurations" (MIRCs), which are computationally derived and have unique psychophysical characteristics, serve as the building blocks of object recognition. We contrasted the BOLD responses elicited by MIRC images, derived from different categories (faces, objects, and places), sub-MIRCs, which are visually similar to MIRCs, but, instead, result in poor recognition and scrambled, unrecognizable images. Stimuli were presented in blocks, and participants indicated yes/no recognition for each image. We confirmed that MIRCs elicited higher recognition performance compared to sub-MIRCs for all three categories. Whereas fMRI activation in early visual cortex for both MIRCs and sub-MIRCs of each category did not differ from that elicited by scrambled images, high-level visual regions exhibited overall greater activation for MIRCs compared to sub-MIRCs or scrambled images. Moreover, MIRCs and sub-MIRCs from each category elicited enhanced activation in corresponding category-selective regions including fusiform face area and occipital face area (faces), lateral occipital cortex (objects), and parahippocampal place area and transverse occipital sulcus (places). These findings reveal the psychological and neural relevance of MIRCs and enable us to make progress in developing a more complete account of object recognition.


Subject(s)
Pattern Recognition, Visual/physiology , Recognition, Psychology/physiology , Visual Cortex/physiology , Adult , Brain/physiology , Brain Mapping , Female , Humans , Magnetic Resonance Imaging , Male , Photic Stimulation , Young Adult
9.
Cognition ; 183: 67-81, 2019 02.
Article in English | MEDLINE | ID: mdl-30419508

ABSTRACT

Rapid developments in the fields of learning and object recognition have been obtained by successfully developing and using methods for learning from a large number of labeled image examples. However, such current methods cannot explain infants' learning of new concepts based on their visual experience, in particular, the ability to learn complex concepts without external guidance, as well as the natural order in which related concepts are acquired. A remarkable example of early visual learning is the category of 'containers' and the notion of 'containment'. Surprisingly, this is one of the earliest spatial relations to be learned, starting already around 3 month of age, and preceding other common relations (e.g., 'support', 'in-between'). In this work we present a model, which explains infants' capacity of learning 'containment' and related concepts by 'just looking', together with their empirical development trajectory. Learning occurs in the model fast and without external guidance, relying only on perceptual processes that are present in the first months of life. Instead of labeled training examples, the system provides its own internal supervision to guide the learning process. We show how the detection of so-called 'paradoxical occlusion' provides natural internal supervision, which guides the system to gradually acquire a range of useful containment-related concepts. Similar mechanisms of using implicit internal supervision can have broad application in other cognitive domains as well as artificial intelligent systems, because they alleviate the need for supplying extensive external supervision, and because they can guide the learning process to extract concepts that are meaningful to the observer, even if they are not by themselves obvious, or salient in the input.


Subject(s)
Child Development/physiology , Learning/physiology , Models, Theoretical , Space Perception/physiology , Visual Perception/physiology , Humans , Infant
10.
PLoS Pathog ; 14(5): e1007044, 2018 05.
Article in English | MEDLINE | ID: mdl-29727445

ABSTRACT

The ability of the Lentivirus HIV-1 to inhibit T-cell activation by its gp41 fusion protein is well documented, yet limited data exists regarding other viral fusion proteins. HIV-1 utilizes membrane binding region of gp41 to inhibit T-cell receptor (TCR) complex activation. Here we examined whether this T-cell suppression strategy is unique to the HIV-1 gp41. We focused on T-cell modulation by the gp21 fusion peptide (FP) of the Human T-lymphotropic Virus 1 (HTLV-1), a Deltaretrovirus that like HIV infects CD4+ T-cells. Using mouse and human in-vitro T-cell models together with in-vivo T-cell hyper activation mouse model, we reveal that HTLV-1's FP inhibits T-cell activation and unlike the HIV FP, bypasses the TCR complex. HTLV FP inhibition induces a decrease in Th1 and an elevation in Th2 responses observed in mRNA, cytokine and transcription factor profiles. Administration of the HTLV FP in a T-cell hyper activation mouse model of multiple sclerosis alleviated symptoms and delayed disease onset. We further pinpointed the modulatory region within HTLV-1's FP to the same region previously identified as the HIV-1 FP active region, suggesting that through convergent evolution both viruses have obtained the ability to modulate T-cells using the same region of their fusion protein. Overall, our findings suggest that fusion protein based T-cell modulation may be a common viral trait.


Subject(s)
HIV Envelope Protein gp41/immunology , Human T-lymphotropic virus 1/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Viral Fusion Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Membrane/metabolism , Cells, Cultured , HIV Infections/immunology , HIV-1/immunology , Humans , Lymphocyte Activation , Membrane Fusion , Mice , Mice, Inbred C57BL , env Gene Products, Human Immunodeficiency Virus/genetics
11.
Arch Womens Ment Health ; 20(1): 139-147, 2017 02.
Article in English | MEDLINE | ID: mdl-27796596

ABSTRACT

Body image disturbances are a prominent feature of eating disorders (EDs). Our aim was to test and evaluate a computerized assessment of body image (CABI), to compare the body image disturbances in different ED types, and to assess the factors affecting body image. The body image of 22 individuals undergoing inpatient treatment with restricting anorexia nervosa (AN-R), 22 with binge/purge AN (AN-B/P), 20 with bulimia nervosa (BN), and 41 healthy controls was assessed using the Contour Drawing Rating Scale (CDRS), the CABI, which simulated the participants' self-image in different levels of weight changes, and the Eating Disorder Inventory-2-Body Dissatisfaction (EDI-2-BD) scale. Severity of depression and anxiety was also assessed. Significant differences were found among the three scales assessing body image, although most of their dimensions differentiated between patients with EDs and controls. Our findings support the use of the CABI in the comparison of body image disturbances in patients with EDs vs. CONTROLS: Moreover, the use of different assessment tools allows for a better understanding of the differences in body image disturbances in different ED types.


Subject(s)
Anorexia Nervosa/psychology , Body Image , Bulimia Nervosa/psychology , Computers , Self Concept , Adolescent , Adult , Anxiety/complications , Anxiety/psychology , Case-Control Studies , Depression/complications , Depression/psychology , Female , Humans , Image Processing, Computer-Assisted , Israel , Severity of Illness Index , Surveys and Questionnaires , Young Adult
12.
Proc Natl Acad Sci U S A ; 113(10): 2744-9, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26884200

ABSTRACT

Discovering the visual features and representations used by the brain to recognize objects is a central problem in the study of vision. Recently, neural network models of visual object recognition, including biological and deep network models, have shown remarkable progress and have begun to rival human performance in some challenging tasks. These models are trained on image examples and learn to extract features and representations and to use them for categorization. It remains unclear, however, whether the representations and learning processes discovered by current models are similar to those used by the human visual system. Here we show, by introducing and using minimal recognizable images, that the human visual system uses features and processes that are not used by current models and that are critical for recognition. We found by psychophysical studies that at the level of minimal recognizable images a minute change in the image can have a drastic effect on recognition, thus identifying features that are critical for the task. Simulations then showed that current models cannot explain this sensitivity to precise feature configurations and, more generally, do not learn to recognize minimal images at a human level. The role of the features shown here is revealed uniquely at the minimal level, where the contribution of each feature is essential. A full understanding of the learning and use of such features will extend our understanding of visual recognition and its cortical mechanisms and will enhance the capacity of computational models to learn from visual experience and to deal with recognition and detailed image interpretation.


Subject(s)
Neural Networks, Computer , Pattern Recognition, Visual/physiology , Vision, Ocular/physiology , Visual Perception/physiology , Brain/physiology , Humans , Models, Neurological , Nerve Net/physiology , Photic Stimulation , Psychophysics/methods , Visual Cortex/physiology , Visual Pathways/physiology
13.
PLoS One ; 10(7): e0130797, 2015.
Article in English | MEDLINE | ID: mdl-26158644

ABSTRACT

Type I interferons are multi-potent cytokines that serve as first line of defense against viruses and other pathogens, posses immunomudolatory functions and elicit a growth inhibitory response. In recent years it has been shown that interferons are also detrimental, for example in lupus, AIDS, tuberculosis and cognitive decline, highlighted the need to develop interferon antagonists. We have previously developed the antagonist IFN-1ant, with much reduced binding to the IFNAR1 receptor and enhanced binding to IFNAR2. Here, we further tune the IFN-1ant by producing three additional antagonists based on IFN-1ant but with altered activity profiles. We show that in all three cases the antiproliferative activity of interferons is blocked and the induction of gene transcription of immunomudolatory and antiproliferative associated genes are substantially decreased. Conversely, each of the new antagonists elicits a different degree of antiviral response, STAT phosphorylation and related gene induction. Two of the new antagonists promote decreased activity in relation to the original IFN-1ant, while one of them promotes increased activity. As we do not know the exact causes of the detrimental effects of IFNs, the four antagonists that were produced and analyzed provide the opportunity to investigate the extent of antagonistic and agonistic activity optimal for a given condition.


Subject(s)
Interferon Type I/genetics , Mutant Proteins/genetics , Mutation , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Encephalomyocarditis virus/drug effects , Gene Expression/drug effects , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Mice, Inbred C57BL , Mutant Proteins/metabolism , Mutant Proteins/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Receptor, Interferon alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Vesicular stomatitis Indiana virus/drug effects
14.
Hum Mol Genet ; 24(11): 3192-205, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25721402

ABSTRACT

We analysed gene expression microarray data from whole blood samples from 228 multiple sclerosis (MS) patients either untreated or treated with one of three alternative commonly used interferon beta (IFNß) disease modifying drugs: Avonex (×1 weekly), Betaseron (every second day) or Rebif (×3 weekly). Patient injections were not timed to coordinate sample collections, thus providing a global transcriptomic profile for each population of patients studied. Three hundred and fifty one genes were significantly differentially expressed by at least one of the IFNß drugs. Despite the different drug sources with distinct injection and dosage protocols, a striking similarity was found in the identity and functional classes of the differentially expressed genes induced. Using the 25 most-upregulated genes, we defined a robust IFNß gene expression signature that quantifies the IFN activation state per blood sample collected irrespective of the type of IFNß therapy. This 25-gene signature also defined basal IFN activation states among untreated MS patients, which differed among individuals but remained relatively constant per patient with time. The maximum drug-induced IFN-activation state was similar for all three drugs despite a 1.7-2.0-fold diminished average effect for Avonex. This and a more erratic effect of Avonex per patient across longitudinal measurements is likely a result of its reduced injection frequency. In summary, we have defined a robust blood-derived type I IFN gene signature from MS patients. This signature could potentially serve to generically quantify the systemic Type I IFN activation status for any other clinical manifestation, inclusive of other autoimmune diseases.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Multiple Sclerosis/blood , Transcriptome/drug effects , Adjuvants, Immunologic/therapeutic use , Humans , Interferon beta-1a/therapeutic use , Interferon beta-1b/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
15.
J Biol Chem ; 289(42): 29014-29, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25193661

ABSTRACT

IFNß is a common therapeutic option to treat multiple sclerosis. It is unique among the family of type I IFNs in that it binds to the interferon receptors with high affinity, conferring exceptional biological properties. We have previously reported the generation of an interferon superagonist (dubbed YNSα8) that is built on the backbone of a low affinity IFNα but modified to exhibit higher receptor affinity than even for IFNß. Here, YNSα8 was fused with a 600-residue hydrophilic, unstructured N-terminal polypeptide chain comprising proline, alanine, and serine (PAS) to prolong its plasma half-life via "PASylation." PAS-YNSα8 exhibited a 10-fold increased half-life in both pharmacodynamic and pharmacokinetic assays in a transgenic mouse model harboring the human receptors, notably without any detectable loss in biological potency or bioavailability. This long-lived superagonist conferred significantly improved protection from MOG35-55-induced experimental autoimmune encephalomyelitis compared with IFNß, despite being injected with a 4-fold less frequency and at an overall 16-fold lower dosage. These data were corroborated by FACS measurements showing a decrease of CD11b(+)/CD45(hi) myeloid lineage cells detectable in the CNS, as well as a decrease in IBA(+) cells in spinal cord sections determined by immunohistochemistry for PAS-YNSα8-treated animals. Importantly, PAS-YNSα8 did not induce antibodies upon repeated administration, and its biological efficacy remained unchanged after 21 days of treatment. A striking correlation between increased levels of CD274 (PD-L1) transcripts from spleen-derived CD4(+) cells and improved clinical response to autoimmune encephalomyelitis was observed, indicating that, at least in this mouse model of multiple sclerosis, CD274 may serve as a biomarker to predict the effectiveness of IFN therapy to treat this complex disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon Type I/agonists , Interferon Type I/pharmacology , Peptides/chemistry , Animals , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Flow Cytometry , Humans , Interferon-beta/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/drug therapy , Protein Engineering/methods , Recombinant Proteins/chemistry , Surface Plasmon Resonance , Treatment Outcome
16.
PLoS One ; 9(1): e84259, 2014.
Article in English | MEDLINE | ID: mdl-24416207

ABSTRACT

We have generated transgenic mice that harbor humanized type I interferon receptors (IFNARs) enabling the study of type I human interferons (Hu-IFN-Is) in mice. These "HyBNAR" (Hybrid IFNAR) mice encode transgenic variants of IFNAR1 and IFNAR2 with the human extracellular domains being fused to transmembrane and cytoplasmic segments of mouse sequence. B16F1 mouse melanoma cells harboring the HyBNAR construct specifically bound Hu-IFN-Is and were rendered sensitive to Hu-IFN-I stimulated anti-proliferation, STAT1 activation and activation of a prototypical IFN-I response gene (MX2). HyBNAR mice were crossed with a transgenic strain expressing the luciferase reporter gene under the control of the IFN-responsive MX2 promoter (MX2-Luciferase). Both the HyBNAR and HyBNAR/MX2-Luciferase mice were responsive to all Hu-IFN-Is tested, inclusive of IFNα2A, IFNß, and a human superagonist termed YNSα8. The mice displayed dose-dependent pharmacodynamic responses to Hu-IFN-I injection, as assessed by measuring the expression of IFN-responsive genes. Our studies also demonstrated a weak activation of endogenous mouse interferon response, especially after high dose administration of Hu-IFNs. In sharp contrast to data published for humans, our pharmacodynamic readouts demonstrate a very short-lived IFN-I response in mice, which is not enhanced by sub-cutaneous (SC) injections in comparison to other administration routes. With algometric differences between humans and mice taken into account, the HyBNAR mice provides a convenient non-primate pre-clinical model to advance the study of human IFN-Is.


Subject(s)
Interferon Type I/metabolism , Animals , Cell Line , Homozygote , Humans , Interferon Type I/pharmacology , Luciferases/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/drug effects , Species Specificity , Transfection , Transgenes
17.
Mol Cell Biol ; 33(4): 800-14, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230268

ABSTRACT

Interferons induce a pleiotropy of responses through binding the same cell surface receptor. Here we investigated the molecular mechanism driving interferon-induced apoptosis. Using a nonbiased small interfering RNA (siRNA) screen, we show that silencing genes whose products are directly engaged in the initiation of interferon signaling completely abrogate the interferon antiproliferative response. Apoptosis-related genes such as the caspase-8, cFLIP, and DR5 genes specifically interfere with interferon-induced apoptosis, which we found to be independent of the activity of death ligands. The one gene for which silencing resulted in the strongest proapoptotic effect upon interferon signaling is the cFLIP gene, where silencing shortened the time of initiation of apoptosis from days to hours and increased dramatically the population of apoptotic cells. Thus, cFLIP serves as a regulator for interferon-induced apoptosis. A shift over time in the balance between cFLIP and caspase-8 results in downstream caspase activation and apoptosis. While gamma interferon (IFN-γ) also causes caspase-8 upregulation, we suggest that it follows a different path to apoptosis.


Subject(s)
Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Interferon Type I/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Enzyme Activation , Gene Expression Regulation , Humans , Interferon-gamma/metabolism , RNA Interference , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
18.
Proc Natl Acad Sci U S A ; 109(44): 18215-20, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23012418

ABSTRACT

Early in development, infants learn to solve visual problems that are highly challenging for current computational methods. We present a model that deals with two fundamental problems in which the gap between computational difficulty and infant learning is particularly striking: learning to recognize hands and learning to recognize gaze direction. The model is shown a stream of natural videos and learns without any supervision to detect human hands by appearance and by context, as well as direction of gaze, in complex natural scenes. The algorithm is guided by an empirically motivated innate mechanism--the detection of "mover" events in dynamic images, which are the events of a moving image region causing a stationary region to move or change after contact. Mover events provide an internal teaching signal, which is shown to be more effective than alternative cues and sufficient for the efficient acquisition of hand and gaze representations. The implications go beyond the specific tasks, by showing how domain-specific "proto concepts" can guide the system to acquire meaningful concepts, which are significant to the observer but statistically inconspicuous in the sensory input.


Subject(s)
Visual Perception , Hand , Humans , Task Performance and Analysis
19.
Mol Cell Biol ; 31(16): 3252-66, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21690295

ABSTRACT

Type I interferons trigger diverse biological effects by binding a common receptor, composed of IFNAR1 and IFNAR2. Intriguingly, while the activation of an antiviral state is common to all cells, antiproliferative activity and apoptosis affect only part of the population, even when cells are stimulated with saturating interferon concentrations. Manipulating receptor expression by different small interfering RNA (siRNA) concentrations reduced the fraction of responsive cells independent of the interferon used, including a newly generated, extremely tight-binding variant. Reduced receptor numbers increased 50% effective concentrations (EC(50)s) for alpha interferon 2 (IFN-α2) but not for the tight-binding variant. A correlation between receptor numbers, STAT activation, and gene induction is observed. Our data suggest that for a given cell, the response is binary (+/-) and dependent on the stochastic expression levels of the receptors on an individual cell. A low number of receptors suffices for antiviral response and is thus a robust feature common to all cells. Conversely, a high number of receptors is required for antiproliferative activity, which allows for fine-tuning on a single-cell level.


Subject(s)
Cell Lineage , Cell Proliferation , Interferon Type I/pharmacology , Receptor, Interferon alpha-beta/genetics , Transcriptional Activation , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , RNA, Small Interfering/pharmacology , Receptor, Interferon alpha-beta/physiology , STAT Transcription Factors/metabolism , Stochastic Processes , Transcriptional Activation/drug effects
20.
Mol Cell Biol ; 29(17): 4778-87, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564411

ABSTRACT

Multiple type I interferons (IFN-alpha/beta) elicit Jak/Stat activation, rapid gene induction, and pleiotropic effects, such as differentiation, antiviral protection, and blocks in proliferation, which are dependent on the IFN subtype and the cellular context. To date, ligand- and receptor-specific molecular determinants underlying IFN-alpha/beta differential activities or potencies have been well characterized. To analyze cellular determinants that impact subtype-specific potency, human fibrosarcoma U5A-derived clones, exhibiting a gradient of IFN sensitivity by virtue of increasing receptor levels, were monitored for Jak/Stat signaling, gene induction, cell cycle lengthening, and apoptosis. In cells with scarce receptors, IFN-beta was more potent than IFN-alpha2 in antiproliferative activity, while the two subtypes were equipotent in all other readouts. Conversely, in cells with abundant receptors, IFN-alpha2 matched or even surpassed IFN-beta in all readouts tested. Our results suggest that the differential activities of the IFN subtypes are dictated not only by the intrinsic ligand/receptor binding kinetics but also by the density of cell surface receptor components.


Subject(s)
Interferon-alpha/metabolism , Interferon-beta/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Line, Tumor , Enzyme Activation , Gene Expression , Humans , Interferon-alpha/genetics , Interferon-beta/genetics , Janus Kinases/genetics , Janus Kinases/metabolism , Receptor, Interferon alpha-beta/genetics , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/physiology , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...