Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Intern Med ; 63(8): 1113-1117, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-37661454

ABSTRACT

A 54-year-old woman with multiple sclerosis treated with interferon-ß (IFN-ß)-1b for 15 years presented with sustained hypertension (240/124 mmHg) and retinal bleeding. She had proteinuria, anemia, thrombocytopenia, elevated serum creatinine levels, and haptoglobin depletion. Intravenous nicardipine stabilized her blood pressure, but her renal function and platelet count deteriorated. The initial disintegrin-like metalloprotease with thrombospondin type 1 motifs 13 (ADAMTS13) activity was 28% of normal without its inhibitor. The subsequent peripheral appearance of schistocytes suggested thrombotic microangiopathy (TMA). After IFN-ß-1b cessation, the platelet count increased, and the blood pressure stabilized. The ADAMTS13 activity normalized, although the creatinine level did not. TMA may develop after the long-term use of IFN-ß without adverse events.


Subject(s)
Hypertension , Multiple Sclerosis , Thrombotic Microangiopathies , Female , Humans , Middle Aged , Interferon beta-1b/adverse effects , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Multiple Sclerosis/chemically induced , Thrombotic Microangiopathies/chemically induced , Interferon-beta/adverse effects , Hypertension/complications
2.
Front Immunol ; 14: 1163739, 2023.
Article in English | MEDLINE | ID: mdl-37025993

ABSTRACT

Aim: To investigate the molecular mechanism underlying the onset of choroidal neovascularization (CNV). Methods: Integrated transcriptomic and proteomic analyses of retinas in mice with laser-induced CNV were performed using RNA sequencing and tandem mass tag. In addition, the laser-treated mice received systemic interferon-ß (IFN-ß) therapy. Measurements of CNV lesions were acquired by the confocal analysis of stained choroidal flat mounts. The proportions of T helper 17 (Th17) cells were determined by flow cytometric analysis. Results: A total of differentially expressed 186 genes (120 up-regulated and 66 down-regulated) and 104 proteins (73 up-regulated and 31 down-regulated) were identified. The gene ontology and KEGG pathway analyses indicated that CNV was mainly associated with immune and inflammatory responses, such as cellular response to IFN-ß and Th17 cell differentiation. Moreover, the key nodes of the protein-protein interaction network mainly involved up-regulated proteins, including alpha A crystallin and fibroblast growth factor 2, and were verified by Western blotting. To confirm the changes in gene expression, real-time quantitative PCR was performed. Furthermore, levels of IFN-ß in both the retina and plasma, as measured by enzyme-linked immunosorbent assay (ELISA), were significantly lower in the CNV group than in the control group. IFN-ß treatment significantly reduced CNV lesion size and promoted the proliferation of Th17 cells in laser-treated mice. Conclusions: This study demonstrates that the occurrence of CNV might be associated with the dysfunction of immune and inflammatory processes and that IFN-ß could serve as a potential therapeutic target.


Subject(s)
Choroidal Neovascularization , Interferon-beta , Mice , Animals , Proteomics , Choroidal Neovascularization/drug therapy , Retina/pathology , Signal Transduction
3.
Cancers (Basel) ; 15(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36980556

ABSTRACT

Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Thus, strategies to mitigate toxicity are important and can revitalize the current standard of care. We previously established that LR-IL-22 protects the intestine from WAI. We now hypothesize that LR-IFN-ß is an effective radiation protector and mitigator and is rapidly cleared from the digestive tract, making it an option for intestinal radioprotection. We report that the gavage of LR-IFN-ß during WAI provides improved intestinal barrier integrity and significantly preserves the numbers of Lgr5+GFP+ intestinal stem cells, improving survival. The rapid clearance of the genetically engineered probiotic from the digestive tract renders it a safe and feasible radiation mitigator. Therefore, the above genetically engineered probiotic is both a feasible and effective radiation mitigator that could potentially revolutionize the management of OC patients. Furthermore, the subsequent addition of platinum/taxane-based chemotherapy to the combination of WAI and LR-IFN-ß should reduce tumor volume while protecting the intestine and should improve the overall survival in OC patients.

4.
Virus Genes ; 57(1): 60-71, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33389635

ABSTRACT

Peste des petits ruminants virus (PPRV) causes an acute and highly contagious disease in domestic and wild small ruminants throughout the world, mainly by invoking immunosuppression in its natural hosts. It has been suggested that the non-structural C protein of PPRV helps in evading host responses but the molecular mechanisms by which it antagonizes the host responses have not been fully characterized. Here, we report the antagonistic effect of PPRV C protein on the expression of interferon-ß (IFN-ß) through both MAVS and RIG-I mediated pathways in vitro. Dual luciferase reporter assay and direct expression of IFN-ß mRNA analysis indicated that PPRV C significantly down regulates IFN-ß via its potential interaction with MAVS and RIG-I signaling molecules. Results further indicated that PPRV C protein significantly suppresses endogenous and exogenous IFN-ß-induced anti-viral effects in PPRV, EMCV and SVS infections in vitro. Moreover, PPRV C protein not only down regulates IFN-ß but also the downstream cytokines of interferon stimulated genes 56 (ISG56), ISG15, C-X-C motif chemokine (CXCL10) and RIG-I mediated activation of IFN promoter elements of ISRE and NF-κB. Further, this study deciphers that PPRV C protein could significantly inhibit the phosphorylation of STAT1 and interferes with the signal transmission in JAK-STAT signaling pathway. Collectively, this study indicates that PPRV C protein is important for innate immune evasion and disease progression.


Subject(s)
Interferon-beta/metabolism , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/metabolism , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chlorocebus aethiops , DEAD Box Protein 58/metabolism , HEK293 Cells , Humans , Receptors, Immunologic/metabolism , Signal Transduction , Vero Cells
5.
J Cell Physiol ; 235(11): 8736-8746, 2020 11.
Article in English | MEDLINE | ID: mdl-32324266

ABSTRACT

Adipose-derived stem cells (ADSCs) are a subset of mesenchymal stem cells that their therapeutic effects in various diseases make them an interesting tool in cell therapy. In the current study, we aimed to overexpress interferon-ß (IFN-ß) and leukemia inhibitory factor (LIF) cytokines in human ADSCs to evaluate the impact of this overexpression on human ADSCs properties. Here, we designed a construct containing IFN-ß and LIF and then, transduced human adipose-derived stem cells (hADSCs) by this construct via a lentiviral vector (PCDH-513B). We assessed the ability of long-term expression of the transgene in transduced cells by western blot analysis and enzyme-linked immunosorbent assay techniques on Days 15, 45, and 75 after transduction. For the evaluation of stem cell properties, flow cytometry and differentiation assays were performed. Finally, the MTT assay was done to assess the proliferation of transduced cells compares to controls. Our results showed high-efficiency transduction with highest expression rates on Day 75 after transduction which were 70 pg/ml for IFN-ß and 77.9 pg/ml for LIF in comparison with 25.60 pg/ml and 27.63 pg/ml, respectively, in untransduced cells (p = .0001). Also, transduced cells expressed a high level of ADSCs surface markers and successfully differentiated into adipocytes, chondrocytes, neural cells, and osteocytes besides the preservation rate of proliferation near untreated cells (p = .88). All in all, we successfully constructed an hADSC population stably overexpressed IFN-ß and LIF cytokines. Considering the IFN-ß and LIF anti-inflammatory and neuroprotective effects as well as immune-regulatory properties of hADSCs, the obtained cells of this study could be subjected for further evaluations in experimental autoimmune encephalomyelitis mice model.


Subject(s)
Adipocytes/metabolism , Interferon-beta/metabolism , Leukemia Inhibitory Factor/metabolism , Mesenchymal Stem Cells/metabolism , Adipose Tissue/cytology , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , Osteocytes/metabolism , Stem Cells/metabolism
6.
Front Immunol ; 10: 1659, 2019.
Article in English | MEDLINE | ID: mdl-31396213

ABSTRACT

Although mesenchymal stromal cells (MSCs) are among the most promising cell sources for cell-based therapies and regenerative medicine, the decline in their function with age due to cellular senescence limits their therapeutic applications. Unveiling the underlying mechanism of MSC senescence is therefore of substantial interest with regard to advancing MSC-based cell therapies. We here show that the induction of human umbilical cord blood-derived MSC (UCB-MSC) senescence causes the predominant upregulation of Toll-like receptor 3 (TLR3). Subsequent TLR3 activation by polyinosinic-polycytidylic acid triggers the prominent features of senescence. Using a clustered regularly interspaced short palindromic repeats/Cas9 library screening system, we identified Janus kinase 1 (JAK1) as the candidate regulatory factor for TLR3-mediated MSC senescence. A JAK1 deficiency blocked the MSC senescence phenotype upon TLR3 activation and TLR3 induction. Targeting the JAK1 pathway using chemical JAK1 inhibitors also significantly suppressed TLR3-mediated MSC senescence. Importantly, we further observed that UCB-MSC senescence is driven by a senescence-associated secretory phenotype (SASP) and that interferon-ß (IFN-ß) is a component of TLR3-dependent SASP, whereby its autocrine actions upregulate TLR3 and suppress cell proliferation. A JAK1 depletion significantly interrupted these effects of IFN-ß, indicating that JAK1 is a signaling mediator linking IFN-ß activity to TLR3 expression and the process of MSC senescence. Collectively, our findings provide new mechanistic insights into UCB-MSC senescence by revealing the role of an autocrine regulatory loop of SASP evoked by TLR3 activation.


Subject(s)
Autocrine Communication/physiology , Cellular Senescence/physiology , Interleukin-6/metabolism , Janus Kinase 1/metabolism , Mesenchymal Stem Cells/metabolism , Toll-Like Receptor 3/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Up-Regulation
7.
Int J Mol Sci ; 18(12)2017 Dec 16.
Article in English | MEDLINE | ID: mdl-29258190

ABSTRACT

Tuberculosis (TB) is a major health threat to the human population worldwide. The etiology of the disease is Mycobacterium tuberculosis (Mtb), a highly successful intracellular pathogen. It has the ability to manipulate the host immune response and to make the intracellular environment suitable for its survival. Many studies have addressed the interactions between the bacteria and the host immune cells as involving many immune mediators and other cellular players. Interferon-ß (IFN-ß) signaling is crucial for inducing the host innate immune response and it is an important determinant in the fate of mycobacterial infection. The role of IFN-ß in protection against viral infections is well established and has been studied for decades, but its role in mycobacterial infections remains much more complicated and debatable. The involvement of IFN-ß in immune evasion mechanisms adopted by Mtb has been an important area of investigation in recent years. These advances have widened our understanding of the pro-bacterial role of IFN-ß in host-pathogen interactions. This pro-bacterial activity of IFN-ß appears to be correlated with its anti-inflammatory characteristics, primarily by antagonizing the production and function of interleukin 1ß (IL-1ß) and interleukin 18 (IL-18) through increased interleukin 10 (IL-10) production and by inhibiting the nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasome. Furthermore, it also fails to provoke a proper T helper 1 (Th1) response and reduces the expression of major histocompatibility complex II (MHC-II) and interferon-γ receptors (IFNGRs). Here we will review some studies to provide a paradigm for the induction, regulation, and role of IFN-ß in mycobacterial infection. Indeed, recent studies suggest that IFN-ß plays a role in Mtb survival in host cells and its downregulation may be a useful therapeutic strategy to control Mtb infection.


Subject(s)
Interferon-beta/metabolism , Tuberculosis/metabolism , Animals , Host-Pathogen Interactions , Humans , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
8.
Virus Res ; 189: 226-34, 2014 Aug 30.
Article in English | MEDLINE | ID: mdl-24911239

ABSTRACT

Type I interferons (IFNs), predominantly IFN-α and ß, play important roles in both innate and adaptive immune responses against viral infections. Porcine reproductive and respiratory syndrome virus (PRRSV) has been recognized to be able to down-regulate the IFN response during in vivo and in vitro infection. In this study, we first analyzed inhibitory effect of each NSP of low pathogenic PRRSV HB-1/3.9 on IFN-ß transcription in MARC-145 cells, and the results showed that the IFN-ß promoter activation could be suppressed by NSP1α, NSP2, NSP1ß, NSP3, NSP4, NSP5 and NSP11. We next confirmed that the inhibitory effect of NSP4 was mainly mediated through suppressing NF-κB activation, whereas not hindering NF-κB phosphorylation and nuclear translocation, and nuclear-localized NSP4 was responsible for inhibiting IFN-ß activation. We further found that the NSP4 of different pathogenic PRRSV strains exhibited differential inhibitory effect on IFN-ß, NF-κB, and IRF3 transcription, and the NSP4 of highly pathogenic (HP)-PRRSV could display more strong inhibitory effect. Finally, we determined that the amino acid at residue 155 in NSP4 contributed to its inhibitory effect for IFN-ß transcription in vitro by altering its subcellular distribution. Our findings suggest that the nucleus-localized NSP4 of PRRSV participates in the modulation of the host type I IFNs system, and also provide novel insight for understanding the pathogenesis of the Chinese HP-PRRSV.


Subject(s)
Host-Pathogen Interactions , Interferon-beta/antagonists & inhibitors , Interferon-beta/biosynthesis , Porcine respiratory and reproductive syndrome virus/immunology , Transcription, Genetic , Viral Nonstructural Proteins/immunology , Animals , Cell Line , DNA Mutational Analysis , Haplorhini
SELECTION OF CITATIONS
SEARCH DETAIL