1,483 results on '"sendai virus"'
Search Results
2. Identification and quantification of defective virus genomes in high throughput sequencing data using DVG-profiler, a novel post-sequence alignment processing algorithm.
- Author
-
Bosma, Trent J., Karagiannis, Konstantinos, Santana-Quintero, Luis, Ilyushina, Natalia, Zagorodnyaya, Tatiana, Petrovskaya, Svetlana, Laassri, Majid, Donnelly, Raymond P., Rubin, Steven, Simonyan, Vahan, and Sauder, Christian J.
- Subjects
- *
VIRAL genomes , *SENDAI virus , *PARAINFLUENZA viruses , *VIRUS research - Abstract
Most viruses are known to spontaneously generate defective viral genomes (DVG) due to errors during replication. These DVGs are subgenomic and contain deletions that render them unable to complete a full replication cycle in the absence of a co-infecting, non-defective helper virus. DVGs, especially of the copyback type, frequently observed with paramyxoviruses, have been recognized to be important triggers of the antiviral innate immune response. DVGs have therefore gained interest for their potential to alter the attenuation and immunogenicity of vaccines. To investigate this potential, accurate identification and quantification of DVGs is essential. Conventional methods, such as RT-PCR, are labor intensive and will only detect primer sequence-specific species. High throughput sequencing (HTS) is much better suited for this undertaking. Here, we present an HTS-based algorithm called DVG-profiler to identify and quantify all DVG sequences in an HTS data set generated from a virus preparation. DVG-profiler identifies DVG breakpoints relative to a reference genome and reports the directionality of each segment from within the same read. The specificity and sensitivity of the algorithm was assessed using both in silico data sets as well as HTS data obtained from parainfluenza virus 5, Sendai virus and mumps virus preparations. HTS data from the latter were also compared with conventional RT-PCR data and with data obtained using an alternative algorithm. The data presented here demonstrate the high specificity, sensitivity, and robustness of DVG-profiler. This algorithm was implemented within an open source cloud-based computing environment for analyzing HTS data. DVG-profiler might prove valuable not only in basic virus research but also in monitoring live attenuated vaccines for DVG content and to assure vaccine lot to lot consistency. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
3. A new mechanism of interferon’s antiviral action: Induction of autophagy, essential for paramyxovirus replication, is inhibited by the interferon stimulated gene, TDRD7.
- Author
-
Subramanian, Gayatri, Kuzmanovic, Teodora, Zhang, Ying, Peter, Cara Beate, Veleeparambil, Manoj, Chakravarti, Ritu, Sen, Ganes C., and Chattopadhyay, Saurabh
- Subjects
- *
INTERFERONS , *AUTOPHAGY , *VIRUS diseases , *PARAMYXOVIRUSES , *SENDAI virus - Abstract
The interferon (IFN) system represents the first line of defense against a wide range of viruses. Virus infection rapidly triggers the transcriptional induction of IFN-β and IFN Stimulated Genes (ISGs), whose protein products act as viral restriction factors by interfering with specific stages of virus life cycle, such as entry, transcription, translation, genome replication, assembly and egress. Here, we report a new mode of action of an ISG, IFN-induced TDRD7 (tudor domain containing 7) inhibited paramyxovirus replication by inhibiting autophagy. TDRD7 was identified as an antiviral gene by a high throughput screen of an ISG shRNA library for blocking IFN’s protective effect against Sendai virus (SeV) replication. The antiviral activity of TDRD7 against SeV, human parainfluenza virus 3 and respiratory syncytial virus was confirmed by its genetic ablation or ectopic expression in several types of mouse and human cells. TDRD7’s antiviral action was mediated by its ability to inhibit autophagy, a cellular catabolic process which was robustly induced by SeV infection and required for its replication. Mechanistic investigation revealed that TDRD7 interfered with the activation of AMP-dependent kinase (AMPK), an enzyme required for initiating autophagy. AMPK activity was required for efficient replication of several paramyxoviruses, as demonstrated by its genetic ablation or inhibition of its activity by TDRD7 or chemical inhibitors. Therefore, our study has identified a new antiviral ISG with a new mode of action. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
4. The antiviral action of the RIG-I induced pathway of apoptosis (RIPA) is enhanced by its ability to degrade Otulin, which deubiquitinates IRF3
- Author
-
Rameez Raja and Ganes C. Sen
- Subjects
Mammals ,Enzyme complex ,biology ,Caspase 3 ,Ubiquitin ,Chemistry ,RIG-I ,viruses ,Ubiquitination ,Apoptosis ,Cell Biology ,biology.organism_classification ,Antiviral Agents ,Article ,Sendai virus ,Cell biology ,Deubiquitinating enzyme ,Proteasome ,Virus Diseases ,biology.protein ,Animals ,IRF3 ,Molecular Biology ,Transcription factor ,Caspase - Abstract
Mammalian innate immune response to virus infection is meditated by many cell-intrinsic pathways. RNA viruses, such as Sendai virus, which replicate in the cytoplasm, trigger the RIG-I-like receptor pathway, which activates the transcription factor, IRF3. Activated IRF3 translocates to the nucleus and induces transcription of the genes which encode interferons, the major antiviral cytokines. Interestingly, IRF3 activates another interferon-independent antiviral pathway, called RIG-I induced pathway of apoptosis (RIPA). For activating RIPA, IRF3 translocates from the cytoplasm to mitochondria. RIPA requires linear polyubiquitination of IRF3 by the enzyme complex, LUBAC; ubiquitinated IRF3 binds to Bax and translocates it to mitochondria causing the release of Cytochrome C, activation of caspases and apoptosis of the infected cell. Here, we report that Otulin, the deubiquitinase that removes linear polyubiquitin chains, inhibits RIPA by deubiquitinating IRF3. Ablation of Otulin expression enhanced RIPA and its overexpression inhibited RIPA. In virus-infected cells, to overcome Otulin-mediated inhibition, RIPA actively degrades Otulin. This degradation required sequential actions of RIPA-activated Caspase 3 and proteasomes. Caspase 3 cleaved Otulin at D31; the D31A mutant was not cleaved at all. The caspase-cleaved fragment was totally degraded by proteasomes, which was preceded by its K48-linked ubiquitination. Mass spectrometric analysis of Otulin identified K64 and K197 as the ubiquitinated residues. Otulin interacted with LUBAC after virus infection and the E3-ubiquitin ligase, HOIP, a component of LUBAC, ubiquitinated Otulin to trigger its proteasome-mediated degradation. To assess the impact of Otulin degradation on RIPA-mediated antiviral action, we expressed, in Otulin-ablated cells, a non-degradable mutant of Otulin, in which D31, K64 and K197 had been mutated. The cells expressing the Otulin mutant were less susceptible to virus-induced apoptosis, because RIPA was less active, and consequently virus replication was more robust. Thus, our study has revealed an important positive feedback loop of RIPA.
- Published
- 2021
- Full Text
- View/download PDF
5. Production of therapeutic iduronate‐2‐sulfatase enzyme with a novel single‐stranded RNA virus vector
- Author
-
Masafumi Onodera, Naomi Yoshida, Emika Kikuchi, Mahito Nakanishi, Ryuichi Mashima, Mari Ohira, Torayuki Okuyama, and Shiori Mizuta
- Subjects
biology ,Iduronic Acid ,Genetic enhancement ,Iduronate-2-sulfatase ,RNA ,Iduronate Sulfatase ,Cell Biology ,biology.organism_classification ,Molecular biology ,Sendai virus ,Virus ,Viral vector ,Genetics ,Animals ,Humans ,RNA Viruses ,Mucopolysaccharidosis type II ,Lysosomes ,Gene ,Mucopolysaccharidosis II - Abstract
The Sendai virus vector has received a lot of attention due to its broad tropism for mammalian cells. As a result of efforts for genetic studies based on a mutant virus, we can now express more than 10 genes of up to 13.5 kilo nucleotides in a single vector with high protein expression efficiency. To prove this benefit, we examined the efficacy of the novel ribonucleic acid (RNA) virus vector harboring the human iduronate-2-sulfatase (IDS) gene with 1,653 base pairs, a causative gene for mucopolysaccharidosis type II, also known as a disorder of lysosomal storage disorders. As expected, this novel RNA vector with the human IDS gene exhibited its marked expression as determined by the expression of enhanced green fluorescent protein and IDS enzyme activity. While these cells exhibited a normal growth rate, the BHK-21 transformant cells stably expressing the human IDS gene persistently generated an active human IDS enzyme extracellularly. The human IDS protein produced failed to be incorporated into the lysosome when cells were pretreated with mannose-6-phosphate, demonstrating that this human IDS enzyme has potential for therapeutic use by cross-correction. These results suggest that our novel RNA vector may be applicable for further clinical settings.
- Published
- 2021
- Full Text
- View/download PDF
6. HSPBP1 facilitates cellular RLR-mediated antiviral response by inhibiting the K48-linked ubiquitination of RIG-I
- Author
-
Jing Li, Ting Ling, Liang-Guo Xu, Tao Xie, Jing-Ping Huang, Ya-Xian Yang, and Sheng-Na Li
- Subjects
0301 basic medicine ,viruses ,Immunology ,chemical and pharmacologic phenomena ,Respirovirus Infections ,Sendai virus ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Receptors, Immunologic ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Gene knockdown ,biology ,Chemistry ,RIG-I ,Binding protein ,Ubiquitination ,virus diseases ,Signal transducing adaptor protein ,RNA ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Immunity, Innate ,Cell biology ,HEK293 Cells ,030104 developmental biology ,DEAD Box Protein 58 ,Signal transduction ,IRF3 ,Signal Transduction ,030215 immunology - Abstract
Retinoic acid-inducible gene I (RIG-I) plays a critical role in the recognition of intracytoplasmic viral RNA. Upon binding to the RNA of invading viruses, the activated RIG-I translocates to mitochondria, where it recruits adapter protein MAVS, causing a series of signaling cascades. In this study, we demonstrated that Hsp70 binding protein 1 (HSPBP1) promotes RIG-I-mediated signal transduction. The overexpression of HSPBP1 can increase the stability of RIG-I protein by inhibiting its K48-linked ubiquitination, and promote the activation of IRF3 and the production of IFN-β induced by Sendai virus. Knockdown and knockout of HSPBP1 leads to down-regulation of virus-induced RIG-I expression, inhibits IRF3 activation, and reduces the production of IFNB1. These results indicate that HSPBP1 positively regulates the antiviral signal pathway induced by inhibiting the K48-linked ubiquitination of RIG-I.
- Published
- 2021
- Full Text
- View/download PDF
7. Control of the induction of type I interferon by Peste des petits ruminants virus.
- Author
-
Sanz Bernardo, Beatriz, Goodbourn, Stephen, and Baron, Michael D.
- Subjects
- *
TYPE I interferons , *PESTE des petits ruminants , *MORBILLIVIRUSES , *MESSENGER RNA , *PROTEIN expression - Abstract
Peste des petits ruminants virus (PPRV) is a morbillivirus that produces clinical disease in goats and sheep. We have studied the induction of interferon-β (IFN-β) following infection of cultured cells with wild-type and vaccine strains of PPRV, and the effects of such infection with PPRV on the induction of IFN-β through both MDA-5 and RIG-I mediated pathways. Using both reporter assays and direct measurement of IFN-β mRNA, we have found that PPRV infection induces IFN-β only weakly and transiently, and the virus can actively block the induction of IFN-β. We have also generated mutant PPRV that lack expression of either of the viral accessory proteins (V&C) to characterize the role of these proteins in IFN-β induction during virus infection. Both PPRV_ΔV and PPRV_ΔC were defective in growth in cell culture, although in different ways. While the PPRV V protein bound to MDA-5 and, to a lesser extent, RIG-I, and over-expression of the V protein inhibited both IFN-β induction pathways, PPRV lacking V protein expression can still block IFN-β induction. In contrast, PPRV C bound to neither MDA-5 nor RIG-I, but PPRV lacking C protein expression lost the ability to block both MDA-5 and RIG-I mediated activation of IFN-β. These results shed new light on the inhibition of the induction of IFN-β by PPRV. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
8. IL-27 LIMITS TYPE 2 IMMUNOPATHOLOGY FOLLOWING PARAINFLUENZA VIRUS INFECTION.
- Author
-
Muallem, Gaia, Wagage, Sagie, Sun, Yan, DeLong, Jonathan H., Valenzuela, Alex, Christian, David A., Harms Pritchard, Gretchen, Fang, Qun, Buza, Elizabeth L., Jain, Deepika, Elloso, M. Merle, López, Carolina B., and Hunter, Christopher A.
- Subjects
- *
IMMUNOPATHOLOGY , *PARAINFLUENZA viruses , *VIRUS diseases , *PARAMYXOVIRUSES , *IMMUNE response , *SENDAI virus , *RESPIRATORY disease risk factors - Abstract
Respiratory paramyxoviruses are important causes of morbidity and mortality, particularly of infants and the elderly. In humans, a T helper (Th)2-biased immune response to these infections is associated with increased disease severity; however, little is known about the endogenous regulators of these responses that may be manipulated to ameliorate pathology. IL-27, a cytokine that regulates Th2 responses, is produced in the lungs during parainfluenza infection, but its role in disease pathogenesis is unknown. To determine whether IL-27 limits the development of pathogenic Th2 responses during paramyxovirus infection, IL-27-deficient or control mice were infected with the murine parainfluenza virus Sendai virus (SeV). Infected IL-27-deficient mice experienced increased weight loss, more severe lung lesions, and decreased survival compared to controls. IL-27 deficiency led to increased pulmonary eosinophils, alternatively activated macrophages (AAMs), and the emergence of Th2 responses. In control mice, IL-27 induced a population of IFN-γ+/IL-10+ CD4+ T cells that was replaced by IFN-γ+/IL-17+ and IFN-γ+/IL-13+ CD4+ T cells in IL-27-deficient mice. CD4+ T cell depletion in IL-27-deficient mice attenuated weight loss and decreased AAMs. Elimination of STAT6 signaling in IL-27-deficient mice reduced Th2 responses and decreased disease severity. These data indicate that endogenous IL-27 limits pathology during parainfluenza virus infection by regulating the quality of CD4+ T cell responses and therefore may have therapeutic potential in paramyxovirus infections. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
9. Reconstruction of Alzheimer’s Disease Cell Model In Vitro via Extracted Peripheral Blood Molecular Cells from a Sporadic Patient
- Author
-
Chengcheng Zhou, Qingguang Wu, Xiaoying Su, Changjun Wang, Hanxu Tan, Sijun Liu, Yuying Zhao, Qi Wang, Peifen Yang, Qiusan Lin, and Shijun Li
- Subjects
Article Subject ,biology ,Cell Biology ,biology.organism_classification ,RC31-1245 ,Embryonic stem cell ,Peripheral blood mononuclear cell ,Molecular biology ,Sendai virus ,In vitro ,law.invention ,SOX2 ,law ,KLF4 ,Induced pluripotent stem cell ,Internal medicine ,Molecular Biology ,Polymerase chain reaction - Abstract
The establishment of human-induced pluripotent stem cell (iPSC) models from sporadic Alzheimer’s disease (sAD) patients is necessary and could potentially benefit research into disease etiology and therapeutic strategies. However, the development of sAD iPSC models is still limited due to the multifactorial nature of the disease. Here, we extracted peripheral blood mononuclear cells (PBMCs) from a patient with sAD and induced them into iPSC by introducing the Sendai virus expressing Oct3/4, Sox2, c-Myc, and Klf4, which were subsequently induced into neural cells to build the cell model of AD. Using alkaline phosphatase staining, immunofluorescence staining, karyotype analysis, reverse transcription-polymerase chain reaction (RT-PCR), and teratoma formation in vitro, we demonstrated that the iPSC derived from PMBCs (PBMC-iPSC) had a normal karyotype and potential to differentiate into three embryonic layers. Immunofluorescence staining and quantitative real-time polymerase chain reaction (qPCR) suggested that PBMC-iPSCs were successfully differentiated into neural cells. Detection of beta-amyloid protein oligomer (AβO), beta-amyloid protein 1-40 (Aβ 1-40), and beta-amyloid protein 1-42 (Aβ 1-42) indicated that the AD cell model was satisfactorily constructed in vitro. In conclusion, this study has successfully generated an AD cell model with pathological features of beta-amyloid peptide deposition using PBMC from a patient with sAD.
- Published
- 2020
- Full Text
- View/download PDF
10. Sustainable Tumor-Suppressive Effect of iPSC-Derived Rejuvenated T Cells Targeting Cervical Cancers
- Author
-
Kazuo Ohara, Miki Ando, Jun Ando, Yumi Sakiyama, Midori Ishii, Ayako Masuda, Tokuko Toyota, Mahito Nakanishi, Tadahiro Honda, Yasuhisa Terao, Norio Komatsu, Manami Ohtaka, and Hiromitsu Nakauchi
- Subjects
Cytotoxicity, Immunologic ,SV40 large T antigen ,cervical cancer ,T-Lymphocytes ,viruses ,medicine.medical_treatment ,Induced Pluripotent Stem Cells ,safer iPSCs ,Uterine Cervical Neoplasms ,T-Cell Antigen Receptor Specificity ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,rejuvenated CTL ,Genetics ,medicine ,Humans ,Cytotoxic T cell ,Vector (molecular biology) ,Cytotoxicity ,Induced pluripotent stem cell ,Molecular Biology ,human papilloma virus type 16 ,030304 developmental biology ,Pharmacology ,Cervical cancer ,0303 health sciences ,biology ,Papillomavirus Infections ,Cell Differentiation ,Oncogene Proteins, Viral ,Immunotherapy ,biology.organism_classification ,medicine.disease ,Sendai virus ,Repressor Proteins ,HPV-CTL ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Original Article ,Female ,T-Lymphocytes, Cytotoxic - Abstract
Immunotherapy utilizing induced pluripotent stem cell (iPSC) technology has great potential. Functionally rejuvenated cytotoxic T lymphocytes (CTLs) can survive long-term as young memory T cells in vivo, with continuous tumor eradication. Banking of iPSCs as an unlimited “off-the-shelf” source of therapeutic T cells may be feasible. To generate safer iPSCs, we reprogrammed human papilloma virus type 16 (HPV16) E6-specific CTLs by Sendai virus vector without cotransduction of SV40 large T antigen. The iPSCs efficiently differentiated into HPV16-specific rejuvenated CTLs that demonstrated robust cytotoxicity against cervical cancer. The tumor-suppressive effect of rejuvenated CTLs was stronger and more persistent than that of original peripheral blood CTLs. These rejuvenated HPV16-specific CTLs provide a sustained tumor-suppressive effect even for epithelial cancers and constitute promising immunotherapy for cervical cancer., Graphical Abstract, In this article, Honda and colleagues show that safer iPSCs can be established from an HPV-CTL clone by Sendai virus vector without cotransduction of SV40 large T antigen. The iPSCs efficiently differentiated rejuvenated CTLs and exhibited a sustained cervical cancer-suppressive effect in vitro and in vivo.
- Published
- 2020
- Full Text
- View/download PDF
11. COVID‐19, cilia, and smell
- Author
-
Ming Li, Wei Li, and Guangshuo Ou
- Subjects
0301 basic medicine ,Anosmia ,viruses ,Viral Nonstructural Proteins ,Biology ,medicine.disease_cause ,Sendai virus ,Severity of Illness Index ,Biochemistry ,SARS‐CoV‐2 ,03 medical and health sciences ,Viewpoint ,0302 clinical medicine ,COVID‐19 ,medicine ,Humans ,Molecular Biology ,Ciliary membrane ,Coronavirus ,Centrosome ,SARS-CoV-2 ,Cilium ,cilia ,COVID-19 ,Methyltransferases ,Cell Biology ,Viral membrane ,Orthomyxoviridae ,biology.organism_classification ,Virology ,Smell ,Viewpoints ,Nasal Mucosa ,030104 developmental biology ,smell loss ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,Respiratory epithelium ,medicine.symptom ,Microtubule-Associated Proteins ,RNA Helicases ,Protein Binding - Abstract
The novel coronavirus SARS‐CoV‐2 is the causative agent of the global coronavirus disease 2019 (COVID‐19) outbreak. In addition to pneumonia, other COVID‐19‐associated symptoms have been reported, including loss of smell (anosmia). However, the connection between infection with coronavirus and anosmia remains enigmatic. It has been reported that defects in olfactory cilia lead to anosmia. In this Viewpoint, we summarize transmission electron microscopic studies of cilia in virus‐infected cells. In the human nasal epithelium, coronavirus infects the ciliated cells and causes deciliation. Research has shown that viruses such as influenza and Sendai attach to the ciliary membrane. The Sendai virus enters cilia by fusing its viral membrane with the ciliary membrane. A recent study on SARS‐CoV‐2–human protein–protein interactions revealed that the viral nonstructural protein Nsp13 interacts with the centrosome components, providing a potential molecular link. The mucociliary escalator removes inhaled pathogenic particles and functions as the first line of protection mechanism against viral infection in the human airway. Thus, future investigation into the virus–cilium interface will help further the battle against COVID‐19., The connection between coronavirus infection and anosmia remains enigmatic. Defects in olfactory cilia lead to anosmia. This viewpoint summarizes transmission electron microscopic studies of cilia in virus‐infected cells: Coronavirus infects the ciliated cells in human nasal epithelium and causes deciliation. Influenza viruses attach to the ciliary membrane, and Sendai virus enters cilia by fusing its viral membrane with the ciliary membrane.
- Published
- 2020
- Full Text
- View/download PDF
12. The interferon-inducible protein TDRD7 inhibits AMP-activated protein kinase and thereby restricts autophagy-independent virus replication
- Author
-
Sukanya Chakravarty, R. Travis Taylor, Sonam Popli, Gayatri Subramanian, Saurabh Chattopadhyay, and Ritu Chakravarti
- Subjects
0301 basic medicine ,viruses ,Immunology ,Herpesvirus 1, Human ,AMP-Activated Protein Kinases ,Virus Replication ,Biochemistry ,Virus ,Mice ,03 medical and health sciences ,Viral entry ,Interferon ,Chlorocebus aethiops ,Autophagy ,medicine ,Animals ,Humans ,Protein kinase A ,Vero Cells ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,AMPK ,Cell Biology ,biology.organism_classification ,Sendai virus ,Cell biology ,030104 developmental biology ,Ribonucleoproteins ,Viral replication ,HeLa Cells ,medicine.drug - Abstract
The interferon system is the first line of defense against virus infection. Recently, using a high-throughput genetic screen of a human interferon-stimulated gene short-hairpin RNA library, we identified a viral restriction factor, TDRD7 (Tudor domain–containing 7). TDRD7 inhibits the paramyxo-/pneumoviruses (e.g. Sendai virus and respiratory syncytial virus) by interfering with the virus-induced cellular autophagy pathway, which these viruses use for their replication. Here, we report that TDRD7 is a viral restriction factor against herpes simplex virus (HSV-1). Using knockdown, knockout, and ectopic expression systems, we demonstrate the anti–HSV-1 activity of TDRD7 in multiple human and mouse cell types. TDRD7 inhibited the virus-activated AMP-activated protein kinase (AMPK), which was essential for HSV-1 replication. Genetic ablation or chemical inhibition of AMPK activity suppressed HSV-1 replication in multiple human and mouse cells. Mechanistically, HSV-1 replication after viral entry depended on AMPK but not on its function in autophagy. The antiviral activity of TDRD7 depended on its ability to inhibit virus-activated AMPK. In summary, our results indicate that the newly identified viral restriction factor TDRD7 inhibits AMPK and thereby blocks HSV-1 replication independently of the autophagy pathway. These findings suggest that AMPK inhibition represents a potential strategy to manage HSV-1 infections.
- Published
- 2020
- Full Text
- View/download PDF
13. PYHIN1 regulates pro-inflammatory cytokine induction rather than innate immune DNA sensing in airway epithelial cells
- Author
-
Davide Massa, Marcin Baran, José A. Bengoechea, and Andrew G. Bowie
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Herpesvirus 1, Human ,Sendai virus ,Biochemistry ,Interferon ,RNA, Small Interfering ,Promoter Regions, Genetic ,innate immunity ,PYHIN proteins ,Nuclear Proteins ,Nucleotidyltransferases ,3. Good health ,Cell biology ,Cytokine ,Stimulator of interferon genes ,Cytokines ,RNA Interference ,Tumor necrosis factor alpha ,pattern recognition receptor (PRR) ,DNA sensing ,medicine.drug ,Immunology ,tumor necrosis factor (TNF) ,Biology ,Cell Line ,Proinflammatory cytokine ,03 medical and health sciences ,Immune system ,medicine ,Humans ,Molecular Biology ,Innate immune system ,030102 biochemistry & molecular biology ,IFI16 ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Membrane Proteins ,Epithelial Cells ,Cell Biology ,interleukin 6 (IL-6) ,Phosphoproteins ,Immunity, Innate ,030104 developmental biology ,DNA, Viral ,airway epithelial cells ,DNA viruses ,cGAS ,STING ,Interleukin-1 - Abstract
Animal cells use pattern-recognition receptors (PRRs) to detect specific pathogens. Pathogen detection mounts an appropriate immune response, including interferon and cytokine induction. The intracellular PRR-signaling pathways that detect DNA viruses have been characterized, particularly in myeloid cells. In these pathways, cGMP-AMP synthase (cGAS) and the pyrin and HIN domain family member (PYHIN) protein interferon-γ-inducible protein 16 (IFI16) detect DNA and signal via stimulator of interferon genes protein (STING). However, although airway epithelial cells are frontline sentinels in detecting pathogens, information on how they respond to DNA viruses is limited, and the roles of PYHIN proteins in these cells are unknown. Here, we examined expression and activities of cGAS, STING, and PYHINs in human lung epithelial cells. A549 epithelial cells, commonly used for RNA-sensing studies, failed to respond to DNA because they lacked STING expression, and ectopic STING expression restored a cGAS-dependent DNA response in these cells. In contrast, NuLi-1 immortalized human bronchial epithelial cells did express STING, which was activated after DNA stimulation and mediated DNA-dependent gene induction. PYHIN1, which like IFI16 has been proposed to be a viral DNA sensor, was the only PYHIN protein expressed in both airway epithelial cell types. However, rather than having a role in DNA sensing, PYHIN1 induced proinflammatory cytokines in response to interleukin-1 (IL-1) or tumor necrosis factor α (TNFα) stimulation. Of note, PYHIN1, via its HIN domain, directly induced IL-6 and TNFα transcription, revealing that PYHIN proteins play a role in proinflammatory gene induction in airway epithelial cells.
- Published
- 2020
- Full Text
- View/download PDF
14. The microRNAs miR-302b and miR-372 regulate mitochondrial metabolism via the SLC25A12 transporter, which controls MAVS-mediated antiviral innate immunity
- Author
-
Keisuke Yaku, Daisuke Kinoshita, Takashi Nakagawa, Takumi Koshiba, and Kai Yasukawa
- Subjects
0301 basic medicine ,Mitochondrion ,Biology ,Mitochondrial Membrane Transport Proteins ,Respirovirus Infections ,Sendai virus ,Biochemistry ,Cell Line ,Proinflammatory cytokine ,03 medical and health sciences ,microRNA ,Humans ,Prohibitin ,Molecular Biology ,Innate immune system ,030102 biochemistry & molecular biology ,RNA ,Cell Biology ,Immunity, Innate ,Mitochondria ,Cell biology ,Solute carrier family ,MicroRNAs ,RNA silencing ,030104 developmental biology ,Host-Pathogen Interactions ,Mitochondrial Membranes - Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that suppress the expression of multiple genes and are involved in numerous biologic functions and disorders, including human diseases. Here, we report that two miRNAs, miR-302b and miR-372, target mitochondrial-mediated antiviral innate immunity by regulating mitochondrial dynamics and metabolic demand. Using human cell lines transfected with the synthetic analog of viral dsRNA, poly(I-C), or challenged with Sendai virus, we found that both miRNAs are up-regulated in the cells late after viral infection and ultimately terminate the production of type I interferons and inflammatory cytokines. We found that miR-302b and miR-372 are involved in dynamin-related protein 1 (DRP1)-dependent mitochondrial fragmentation and disrupt mitochondrial metabolism by attenuating solute carrier family 25 member 12 (SLC25A12), a member of the SLC25 family. Neutralizing the effects of the two miRNAs through specific inhibitors re-established the mitochondrial dynamics and the antiviral responses. We found that SLC25A12 contributes to regulating the antiviral response by inducing mitochondrial-related metabolite changes in the organelle. Structure-function analysis indicated that SLC25A12, as part of a prohibitin complex, associates with the mitochondrial antiviral-signaling protein in mitochondria, providing structural insight into the regulation of the mitochondrial-mediated antiviral response. Our results contribute to the understanding of how miRNAs modulate the innate immune response by altering mitochondrial dynamics and metabolic demand. Manipulating the activities of miR-302b and miR-372 may be a potential therapeutic approach to target RNA viruses.
- Published
- 2020
- Full Text
- View/download PDF
15. Molecular cloning and functional characterization of duck Janus kinase 1
- Author
-
Yaqian Li, Hui Jin, Huijun Zheng, Peng Zhou, Dejian Liu, and Rui Luo
- Subjects
0301 basic medicine ,Small interfering RNA ,animal structures ,biology ,Janus kinase 1 ,Activator (genetics) ,viruses ,Immunology ,Janus Kinase 1 ,biology.organism_classification ,Protein kinase R ,Molecular biology ,Immunity, Innate ,Sendai virus ,Avian Proteins ,03 medical and health sciences ,Ducks ,030104 developmental biology ,0302 clinical medicine ,Transcription (biology) ,Viperin ,Animals ,Signal transduction ,Molecular Biology ,030215 immunology - Abstract
Janus kinase 1 (JAK1) is a member of JAK family of non-receptor protein tyrosine kinases that plays critical roles in transducing cytokine signals via JAK-signal transducer and activator of transcription (STAT) signaling pathway. The importance of JAK1 in innate immunity has been well-studied in mammals and fish, yet in avian remains largely unknown. Here, we cloned the full-length of the duck JAK1 (duJAK1) gene for the first time. DuJAK1 encoded a protein of 1152 amino acids and possessed high amino acid identity with goose and budgerigar JAK1s. The duJAK1 was expressed in all detected tissues, especially high in the thymus and bursa of Fabricius. Overexpression of duJAK1 significantly activated ISRE promoter activity and induced duck viperin, 2', 5'-OAS, MX, PKR and ZAP expression. Knockdown of duJAK1 by small interfering RNA significantly inhibited duck Tembusu virus (DTMUV)-, duck Enteritis virus (DEV)-, poly (I:C)-, poly (dA:dT)- or Sendai virus (SeV)-induced ISRE promoter activation. Furthermore, duJAK1 exhibited antiviral activity against DTMUV infection. These results will help us understand the function of JAK family proteins in duck antiviral immunity.
- Published
- 2020
- Full Text
- View/download PDF
16. Airway Surface Liquid Has Innate Antiviral Activity That Is Reduced in Cystic Fibrosis
- Author
-
Ultan F. Power, Mahmoud H. Abou Alaiwa, Paul B. McCray, Steven M. Varga, Abigail R. Berkebile, and Jennifer A. Bartlett
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Cystic Fibrosis ,Swine ,Respiratory System ,Clinical Biochemistry ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Respiratory Mucosa ,Antiviral Agents ,Cystic fibrosis ,Virus ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Extracellular ,medicine ,Humans ,Animals ,Respiratory system ,Lung ,Molecular Biology ,Original Research ,biology ,business.industry ,Editorials ,Cell Biology ,respiratory system ,Hydrogen-Ion Concentration ,medicine.disease ,biology.organism_classification ,Immunity, Innate ,In vitro ,Sendai virus ,Body Fluids ,Trachea ,030104 developmental biology ,030228 respiratory system ,Virus Diseases ,Viruses ,Immunology ,medicine.symptom ,business - Abstract
Although chronic bacterial infections and inflammation are associated with progressive lung disease in patients with cystic fibrosis (CF), much less is known regarding the contributions of respiratory viral infections to this process. Clinical studies suggest that antiviral host defenses may be compromised in individuals with CF, and CF airway epithelia exhibit impaired antiviral responses in vitro. Here, we used the CF pig model to test the hypothesis that the antiviral activity of respiratory secretions is reduced in CF. We developed an in vitro assay to measure the innate antiviral activity present in airway surface liquid (ASL) from CF and non-CF pigs. We found that tracheal and nasal ASL from newborn non-CF pigs exhibited dose-dependent inhibitory activity against several enveloped and encapsidated viruses, including Sendai virus, respiratory syncytial virus, influenza A, and adenovirus. Importantly, we found that the anti–Sendai virus activity of nasal ASL from newborn CF pigs was significantly diminished relative to non-CF littermate controls. This diminution of extracellular antiviral defenses appears to be driven, at least in part, by the differences in pH between CF and non-CF ASL. These data highlight the novel antiviral properties of native airway secretions and suggest the possibility that defects in extracellular antiviral defenses contribute to CF pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
17. Non-transmissible MV Vector with Segmented RNA Genome Establishes Different Types of iPSCs from Hematopoietic Cells
- Author
-
Yoshiyuki Yamaguchi, Makoto Takeda, Ryo Kurita, Takafumi Hiramoto, Hiroyuki Sasaki, Kota Inoue, Akihide Ryo, Yasushi Soda, Shinji Okano, Kenji Ichiyanagi, Atsushi Muraguchi, Hiroshi Kohara, Shohei Miyamoto, Hidehiro Toh, Yasuki Hijikata, Hiroshi Hamana, Hiroyuki Kishi, Maino Tahara, Jiyuan Liao, Kenzaburo Tani, Yoshie Miura, and Yoshinao Oda
- Subjects
Male ,T-Lymphocytes ,Genetic enhancement ,Transgene ,Genetic Vectors ,Induced Pluripotent Stem Cells ,Blood Donors ,Genome, Viral ,Biology ,Sendai virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genome editing ,Mice, Inbred NOD ,Transduction, Genetic ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Transgenes ,Progenitor cell ,Induced pluripotent stem cell ,Molecular Biology ,Gene ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Hematopoietic stem cell ,Cell Differentiation ,Genetic Therapy ,Fibroblasts ,Cellular Reprogramming ,Hematopoietic Stem Cells ,Cell biology ,Haematopoiesis ,HEK293 Cells ,medicine.anatomical_structure ,Measles virus ,030220 oncology & carcinogenesis ,Heterografts ,RNA, Viral ,Molecular Medicine ,Original Article - Abstract
Recent advances in gene therapy technologies have enabled the treatment of congenital disorders and cancers and facilitated the development of innovative methods, including induced pluripotent stem cell (iPSC) production and genome editing. We recently developed a novel non-transmissible and non-integrating measles virus (MV) vector capable of transferring multiple genes simultaneously into a wide range of cells through the CD46 and CD150 receptors. The MV vector expresses four genes for iPSC generation and the GFP gene for a period of time sufficient to establish iPSCs from human fibroblasts as well as peripheral blood T cells. The transgenes were expressed differentially depending on their gene order in the vector. Human hematopoietic stem/progenitor cells were directly and efficiently reprogrammed to naive-like cells that could proliferate and differentiate into primed iPSCs by the same method used to establish primed iPSCs from other cell types. The novel MV vector has several advantages for establishing iPSCs and potential future applications in gene therapy.
- Published
- 2020
- Full Text
- View/download PDF
18. Kinetic analysis of paramyxovirus-sialoglycan receptor interactions reveals virion motility
- Author
-
Xuesheng Wu, Maite Goebbels, Lemeng Chao, Tom Wennekes, Frank J. M. van Kuppeveld, Erik de Vries, Cornelis A. M. de Haan, Afd Chemical Biology and Drug Discovery, and Virologie
- Subjects
Sialic-acid linkages ,Immunology ,Newcastle-disease virus ,Binding-site ,Human parainfluenza virus ,Tropism ,Sendai virus ,Microbiology ,Virology ,Specificity ,Genetics ,Parasitology ,Influenza-a ,Molecular Biology ,Cellular receptors ,Hemagglutinin-neuraminidase protein - Abstract
Many viruses initiate infection by binding to sialoglycan receptors at the cell surface. Binding to such receptors comes at a cost, however, as the sheer abundance of sialoglycans e.g. in mucus, may immobilize virions to non-functional decoy receptors. As a solution, sialoglycan-binding as well as sialoglycan-cleavage activities are often present in these viruses, which for paramyxoviruses are combined in the hemagglutinin-neuraminidase (HN) protein. The dynamic interactions of sialoglycan-binding paramyxoviruses with their receptors are thought to be key determinants of species tropism, replication and pathogenesis. Here we used biolayer interferometry to perform kinetic analyses of receptor interactions of animal and human paramyxoviruses (Newcastle disease virus, Sendai virus, and human parainfluenza virus 3). We show that these viruses display strikingly different receptor interaction dynamics, which correlated with their receptor-binding and -cleavage activities and the presence of a second sialic acid binding site. Virion binding was followed by sialidase-driven release, during which virions cleaved sialoglycans until a virus-specific density was reached, which was largely independent of virion concentration. Sialidase-driven virion release was furthermore shown to be a cooperative process and to be affected by pH. We propose that paramyxoviruses display sialidase-driven virion motility on a receptor-coated surface, until a threshold receptor density is reached at which virions start to dissociate. Similar motility has previously been observed for influenza viruses and is likely to also apply to sialoglycan-interacting embecoviruses. Analysis of the balance between receptor-binding and -cleavage increases our understanding of host species tropism determinants and zoonotic potential of viruses.
- Published
- 2023
- Full Text
- View/download PDF
19. Novel Strategy to Control Transgene Expression Mediated by a Sendai Virus-Based Vector Using a Nonstructural C Protein and Endogenous MicroRNAs.
- Author
-
Sano, Masayuki, Iijima, Minoru, Ohtaka, Manami, and Nakanishi, Mahito
- Subjects
- *
TRANSGENE expression , *SENDAI virus , *VIRAL replication , *NUCLEOTIDE sequence , *GENETIC vectors , *GENETIC transformation - Abstract
Tissue-specific control of gene expression is an invaluable tool for studying various biological processes and medical applications. Efficient regulatory systems have been utilized to control transgene expression in various types of DNA viral or integrating viral vectors. However, existing regulatory systems are difficult to transfer into negative-strand RNA virus vector platforms because of significant differences in their transcriptional machineries. In this study, we developed a novel strategy for regulating transgene expression mediated by a cytoplasmic RNA vector based on a replication-defective and persistent Sendai virus (SeVdp). Because of the capacity of Sendai virus (SeV) nonstructural C proteins to specifically inhibit viral RNA synthesis, overexpression of C protein significantly reduced transgene expression mediated by SeVdp vectors. We found that SeV C overexpression concomitantly reduced SeVdp mRNA levels and genomic RNA synthesis. To control C expression, target sequences for an endogenous microRNA were incorporated into the 3′ untranslated region of the C genes. Incorporation of target sequences for miR-21 into the SeVdp vector restored transgene expression in HeLa cells by decreasing C expression. Furthermore, the SeVdp vector containing target sequences for let-7a enabled cell-specific control of transgene expression in human fibroblasts and induced pluripotent stem cells. Our findings demonstrate that SeV C can be used as an effective regulator for controlling transgene expression. This strategy will contribute to efficient and less toxic SeVdp-mediated gene transfer in various biological applications. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
20. Clinical Evaluation of a Single-Tube Multiple RT-PCR Assay for the Detection of 13 Common Virus Types/Subtypes Associated with Acute Respiratory Infection.
- Author
-
Zhang, Dan, Feng, Zhishan, Zhao, Mengchuan, Wang, Hao, Wang, Le, Yang, Shuo, Li, Guixia, Lu, Li, and Ma, Xuejun
- Subjects
- *
RESPIRATORY syncytial virus infections , *REVERSE transcriptase polymerase chain reaction , *CAUSES of death , *INFANT mortality , *INFANT diseases , *SENDAI virus , *ADULT respiratory distress syndrome - Abstract
Respiratory viruses are among the most important causes of human morbidity and mortality worldwide, especially for infants and young children. In the past years, a few commercial multiplex RT-PCR assays have been used to detect respiratory viruses in spite of the high cost. In the present study, an improved single-tube multiplex reverse transcription PCR assay for simultaneous detection of 13 respiratory viruses was evaluated and compared with a previously reported two-tube assay as the reference method using clinical nasopharyngeal aspirates samples. Of 310 prospectively tested respiratory specimens selected from children hospitalized with acute respiratory illness, 226 (72.90%, 226/310) and 214 (69.03%, 214/310) positive for one or more viruses were identified by the single-tube and the two-tube assays, respectively, with combined test results showing good concordance (Kappa value = 0.874). Individually, the single-tube assay for adenovirus (Adv), human metapneumovirus (HMPV), human rhinovirus (HRV), parainfluenza virus type 1 (PIV1), parainfluenza virus type 3 (PIV3) and parainfluenza virus type 4 (PIV4) showed the significantly superior sensitivities to those of the two-tube assay. No false positives were found. In conclusion, our results demonstrates the one-tube assay revealed significant improvements over the two-tube assay in terms of the better sensitivity, more accurate quality control, less nonspecific amplification, more cost-effective and shorter turn-around time and will be a valuable tool for routine surveillance of respiratory virus infection in China. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
21. Targeted gene correction and functional recovery in achondroplasia patient-derived iPSCs
- Author
-
Fang Luo, Yundong Li, Wenyuan Wang, Huan Zou, Mingfeng Guan, and Yiren Qin
- Subjects
Male ,medicine.medical_specialty ,Medicine (General) ,Induced Pluripotent Stem Cells ,Medicine (miscellaneous) ,Dwarfism ,iPSCs ,QD415-436 ,Biology ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Achondroplasia ,R5-920 ,Internal medicine ,medicine ,Humans ,Induced pluripotent stem cell ,Gene ,Gene Editing ,Mutation ,Point mutation ,Research ,Gene correction ,Cell Differentiation ,Cell Biology ,Fibroblast growth factor receptor 3 ,biology.organism_classification ,medicine.disease ,Molecular biology ,Sendai virus ,Endocrinology ,Cell culture ,Molecular Medicine ,Female ,Stem cell ,CRISPR-Cas9 ,Targeted Gene Repair - Abstract
Achondroplasia (ACH) is the most common genetic form of dwarfism and belong to dominant monogenetic disorder (MGD) caused by a gain-of-function point mutation in the transmembrane region of fibroblast growth factor receptor 3 (FGFR3). There are currently two mutation sites reported – Gly380Arg and Gly375Cys, the former occupies the vast majority of ACH patients. Homozygous ACH patients have a much more severe phenotype and rarely survive, so most of the ACH patients seen are heterozygous mutations. Like many other MGDs, there are no effective therapeutic methods for ACH even though the cause of the mutation has been found. With the rapid development of stem cell biology and gene-editing technology, the research of ACH is not only important, but also provides theoretical and experimental basis for the investigation of other MGDs. In this study, we generated non-integrated induced pluripotent stem cell (iPSC) lines from ACH patient skin and urine of Gly380Arg mutation. Via powerful gene-editing tool – CRISPR-Cas9, we successfully obtained 2 completely corrected knock-in cell lines. They not only expressed pluripotent markers, maintained normal chromosomal number and structure, but also no off-target indels were identified. This is not only the first time to obtain ACH iPSCs using the most readily available biopsy sample – urine, the first to obtain non-integrated ACH iPSCs, but also is the first to perform gene correction in ACH research. Our study may provide important theoretical and experimental basis for the next step of stem cell research and treatment of ACH in the future.
- Published
- 2021
22. Quantifying dose-, strain-, and tissue-specific kinetics of parainfluenza virus infection
- Author
-
Lubna Pinky, Charles J. Russell, Crystal W. Burke, and Amber M. Smith
- Subjects
Viral Diseases ,Luminescence ,Pulmonology ,Physiology ,Respiratory System ,Viral Upper Respiratory Tract Infection ,Sendai virus ,Mice ,Medical Conditions ,Nasopharynx ,Medicine and Health Sciences ,Biology (General) ,Luciferases ,Lung ,Respiratory Tract Infections ,Infectivity ,Paramyxoviridae Infections ,Ecology ,Physics ,Electromagnetic Radiation ,Respiratory infection ,Viral Load ,Trachea ,Human Parainfluenza Virus ,Lower Respiratory Tract Infections ,medicine.anatomical_structure ,Infectious Diseases ,Computational Theory and Mathematics ,Modeling and Simulation ,Host-Pathogen Interactions ,Physical Sciences ,Anatomy ,Bioluminescence ,Viral load ,Research Article ,QH301-705.5 ,Biology ,Microbiology ,Virus ,Cellular and Molecular Neuroscience ,Respiratory Disorders ,Virology ,Genetics ,medicine ,Upper Respiratory Tract Infections ,Animals ,Humans ,Respiratory Physiology ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Biology and Life Sciences ,Respiratory Infections ,Tissue tropism ,Pharynx ,Digestive System ,Viral Transmission and Infection ,Respiratory tract - Abstract
Human parainfluenza viruses (HPIVs) are a leading cause of acute respiratory infection hospitalization in children, yet little is known about how dose, strain, tissue tropism, and individual heterogeneity affects the processes driving growth and clearance kinetics. Longitudinal measurements are possible by using reporter Sendai viruses, the murine counterpart of HPIV 1, that express luciferase, where the insertion location yields a wild-type (rSeV-luc(M-F*)) or attenuated (rSeV-luc(P-M)) phenotype. Bioluminescence from individual animals suggests that there is a rapid increase in expression followed by a peak, biphasic clearance, and resolution. However, these kinetics vary between individuals and with dose, strain, and whether the infection was initiated in the upper and/or lower respiratory tract. To quantify the differences, we translated the bioluminescence measurements from the nasopharynx, trachea, and lung into viral loads and used a mathematical model together a nonlinear mixed effects approach to define the mechanisms distinguishing each scenario. The results confirmed a higher rate of virus production with the rSeV-luc(M-F*) virus compared to its attenuated counterpart, and suggested that low doses result in disproportionately fewer infected cells. The analyses indicated faster infectivity and infected cell clearance rates in the lung and that higher viral doses, and concomitantly higher infected cell numbers, resulted in more rapid clearance. This parameter was also highly variable amongst individuals, which was particularly evident during infection in the lung. These critical differences provide important insight into distinct HPIV dynamics, and show how bioluminescence data can be combined with quantitative analyses to dissect host-, virus-, and dose-dependent effects., Author summary Human parainfluenza viruses (HPIVs) cause acute respiratory infections and can lead to the hospitalization of children. HPIV infection severity may vary due to dose, strain, patient, and whether the infection initiates within the upper or lower respiratory tract. There is a need to determine how the rates of virus spread and clearance change in different infection scenarios in order to better understand varying clinical manifestations. The significance of our research is in identifying the dominant mechanisms driving strain-, dose-, and tissue-specific HPIV infection kinetics, and in pairing bioluminescence data with quantitative analyses to determine how the same virus can yield patient-specific outcomes. This work enhances our understanding of HPIV infection and broadens our knowledge viral dynamics in the upper and lower respiratory tracts.
- Published
- 2021
23. Establishment of an arrhythmogenic right ventricular cardiomyopathy derived iPSC cell line (USFi004-A) carrying a heterozygous mutation in PKP2 (c.1799delA)
- Author
-
Alexander C. Bertalovitz, Jiajia Yang, Mariana Burgos Angulo, Thomas V. McDonald, and Eva Samal
- Subjects
0301 basic medicine ,QH301-705.5 ,Induced Pluripotent Stem Cells ,Peripheral blood mononuclear cell ,Right ventricular cardiomyopathy ,Cell Line ,Kruppel-Like Factor 4 ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Expressivity (genetics) ,Biology (General) ,Induced pluripotent stem cell ,Gene ,Arrhythmogenic Right Ventricular Dysplasia ,biology ,Karyotype ,Cell Biology ,General Medicine ,biology.organism_classification ,Penetrance ,Molecular biology ,Sendai virus ,030104 developmental biology ,Mutation ,Leukocytes, Mononuclear ,Plakophilins ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an autosomal dominant inherited disease, with variable penetrance and expressivity. Currently, more than 14 different genetic loci have been reported for ARVC, the majority being desmosomal genes like Plakophilin-2 (PKP2). Here, we generated an iPSC cell line bearing a pathogenic heterozygous mutation in PKP2 (c.1799delA) from a patient affected by ARVC. Peripheral blood mononuclear cells were reprogrammed by Sendai virus vectors encoding KOS, KLF4, and c-MYC. Derived iPSCs expressed pluripotent markers, showed intact karyotype and demonstrated the ability to differentiate into three germ layers.
- Published
- 2021
24. The RNA binding protein Quaking represses host interferon response by downregulating MAVS
- Author
-
W. Samuel Fagg, Vanessa Chuo, Julien Pompon, Shelton S. Bradrick, Mariano A. Garcia-Blanco, and Kuo-Chieh Liao
- Subjects
RNA-binding protein ,Biology ,Respirovirus Infections ,Sendai virus ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Transcription (biology) ,Interferon ,medicine ,Humans ,Phosphorylation ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,0303 health sciences ,RNA-Binding Proteins ,MDA5 ,Cell Biology ,Cell biology ,Poly I-C ,Gene Expression Regulation ,A549 Cells ,030220 oncology & carcinogenesis ,Monocyte differentiation ,Host-Pathogen Interactions ,Interferon Type I ,Interferon Regulatory Factor-3 ,CRISPR-Cas Systems ,IRF3 ,Research Paper ,medicine.drug - Abstract
Quaking (QKI) is an RNA-binding protein (RBP) involved in multiple aspects of RNA metabolism and many biological processes. Despite a known immune function in regulating monocyte differentiation and inflammatory responses, the degree to which QKI regulates the host interferon (IFN) response remains poorly characterized. Here we show that QKI ablation enhances poly(I:C) and viral infection-induced IFNβ transcription. Characterization of IFN-related signalling cascades reveals that QKI knockout results in higher levels of IRF3 phosphorylation. Interestingly, complementation with QKI-5 isoform alone is sufficient to rescue this phenotype and reduce IRF3 phosphorylation. Further analysis shows that MAVS, but not RIG-I or MDA5, is robustly upregulated in the absence of QKI, suggesting that QKI downregulates MAVS and thus represses the host IFN response. As expected, MAVS depletion reduces IFNβ activation and knockout of MAVS in the QKI knockout cells completely abolishes IFNβ induction. Consistently, ectopic expression of RIG-I activates stronger IFNβ induction via MAVS-IRF3 pathway in the absence of QKI. Collectively, these findings demonstrate a novel role for QKI in negatively regulating host IFN response by reducing MAVS levels.
- Published
- 2019
- Full Text
- View/download PDF
25. Respirovirus C protein inhibits activation of type I interferon receptor‐associated kinases to block JAK‐STAT signaling
- Author
-
Miki Kohno, Mayu Yamaguchi, Yoshinori Kitagawa, Bin Gotoh, Madoka Sakai, and Masae Itoh
- Subjects
food.ingredient ,Protein subunit ,Biophysics ,Receptor, Interferon alpha-beta ,Respirovirus ,Sendai virus ,Biochemistry ,Cell Line ,JAK-STAT pathway ,Viral Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,food ,Structural Biology ,Interferon ,Genetics ,medicine ,Humans ,Phosphorylation ,Molecular Biology ,030304 developmental biology ,TYK2 Kinase ,0303 health sciences ,respirovirus ,biology ,Kinase ,030302 biochemistry & molecular biology ,Tyrosine phosphorylation ,Janus Kinase 1 ,interferon ,Cell Biology ,C protein ,TYK2 ,biology.organism_classification ,Cell biology ,STAT Transcription Factors ,HEK293 Cells ,JAK1 ,chemistry ,Tyrosine kinase 2 ,Mutation ,Signal Transduction ,medicine.drug - Abstract
Respirovirus C protein blocks the type I interferon (IFN)-stimulated activation of the JAK-STAT pathway. It has been reported that C protein inhibits IFN-α-stimulated tyrosine phosphorylation of STATs, but the underlying mechanism is poorly understood. Here, we show that the C protein of Sendai virus (SeV), a member of the Respirovirus genus, binds to the IFN receptor subunit IFN-α/β receptor subunit (IFNAR)2 and inhibits IFN-α-stimulated tyrosine phosphorylation of the upstream receptor-associated kinases, JAK1 and TYK2. Analysis of various SeV C mutant (Cm) proteins demonstrates the importance of the inhibitory effect on receptor-associated kinase phosphorylation for blockade of JAK-STAT signaling. Furthermore, this inhibitory effect and the IFNAR2 binding capacity are observed for all the respirovirus C proteins examined. Our results suggest that respirovirus C protein inhibits activation of the receptor-associated kinases JAK1 and TYK2 possibly through interaction with IFNAR2.
- Published
- 2019
- Full Text
- View/download PDF
26. A Digital Method to Quantify Type I Interferon
- Author
-
Estanislao Nistal-Villán, Susana Esteban-Rubio, Jesús Presa, Sergio Rius-Rocabert, and Angel Ayuso-Sacido
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Biology ,Sendai virus ,Green fluorescent protein ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Bioassay ,030212 general & internal medicine ,Vero Cells ,Cells, Cultured ,Cell Biology ,Molecular biology ,Effective dose (pharmacology) ,Recombinant Proteins ,030104 developmental biology ,Cytokine ,Viral replication ,Lytic cycle ,Cell culture ,Interferon Type I ,Biological Assay ,medicine.drug - Abstract
Interferon (IFN), the first ever-described cytokine, has a potent activity against viruses. Soon since its discovery, quantification of IFN has been an important issue. Most of the traditional methods to measure IFN biological activity rely on indirect methods that quantify dyes retained by IFN-protected cells against a lytic virus, or by techniques that indirectly quantify viral replication by measuring the expression level of viral-encoded reporter proteins such as the green fluorescent protein (GFP). In both cases, the IFN units are determined by the quantification of an effective dose 50, defined as the IFN dose that prevents 50% cell death of 50% reduction of the maximal amount of GFP intensity. In this study we propose the use of an alternative approach to measure IFN activity by calculating the minimal IFN dose 50 as the amount of IFN able to completely protect 50% of the cells from infection measured by the total absence of virus-dependent GFP signal in a cell culture plate. This sensitive approach could be used to easily quantify the Z value to determine IFN bioassay robustness. We believe that this approximation could be interesting to be considered by the IFN community.
- Published
- 2019
- Full Text
- View/download PDF
27. Porcine deltacoronavirus nsp15 antagonizes interferon-β production independently of its endoribonuclease activity
- Author
-
Liurong Fang, Xiaorong Liu, Yingying Hong, Puxian Fang, Xinyu Zhu, Guiqing Peng, Shaobo Xiao, and Dang Wang
- Subjects
0301 basic medicine ,Swine ,viruses ,Endoribonuclease activity ,Immunology ,Endoribonuclease ,Viral Nonstructural Proteins ,Biology ,Virus Replication ,medicine.disease_cause ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,Endoribonucleases ,medicine ,Animals ,Humans ,Interferon production ,Molecular Biology ,Transcription factor ,Coronavirus ,Swine Diseases ,NF-kappa B ,Porcine deltacoronavirus ,Interferon-beta ,RNA-Dependent RNA Polymerase ,biology.organism_classification ,Sendai virus ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Viral replication ,Interferon Type I ,Nonstructural protein 15 (nsp15) ,Coronavirus Infections ,IRF3 ,Signal Transduction ,030215 immunology ,medicine.drug - Abstract
Highlights • PDCoV nsp15 antagonizes IFN-β production. • PDCoV nsp15 suppresses NF-κB activation other than IRF3 activation. • The endoribonuclease activity is not essential for PDCoV nsp15 to antagonize IFN-β., Porcine deltacoronavirus (PDCoV) is an emerging swine coronavirus causing diarrhea and intestinal damage in nursing piglets. Previous work showed that PDCoV infection inhibits type I interferon (IFN) production. To further identify and characterize the PDCoV-encoded IFN antagonists will broaden our understanding of its pathogenesis. Nonstructural protein 15 (nsp15) encodes an endoribonuclease that is highly conserved among vertebrate nidoviruses (coronaviruses and arteriviruses) and plays a critical role in viral replication and transcription. Here, we found that PDCoV nsp15 significantly inhibits Sendai virus (SEV)-induced IFN-β production. PDCoV nsp15 disrupts the phosphorylation and nuclear translocation of NF-κB p65 subunit, but not antagonizes the activation of transcription factor IRF3. Interestingly, site-directed mutagenesis found that PDCoV nsp15 mutants (H129A, H234A, K269A) lacking endoribonuclease activity also suppress SEV-induced IFN-β production and NF-κB activation, suggesting that the endoribonuclease activity is not required for its ability to antagonize IFN-β production. Taken together, our results demonstrate that PDCoV nsp15 is an IFN antagonist and it inhibits interferon-β production via an endoribonuclease activity-independent mechanism.
- Published
- 2019
- Full Text
- View/download PDF
28. Porcine deltacoronavirus nucleocapsid protein antagonizes IFN-β production by impairing dsRNA and PACT binding to RIG-I
- Author
-
Jun Chen, Liurong Fang, Jie Ren, Dang Wang, Zhen Ding, Shaobo Xiao, Puxian Fang, Guiqing Peng, Qi Peng, and Mohan Wang
- Subjects
Swine ,viruses ,medicine.disease_cause ,Sendai virus ,RIG-I ,03 medical and health sciences ,Interferon ,Virology ,Genetics ,Viral structural protein ,medicine ,Animals ,Humans ,Receptors, Immunologic ,Nucleocapsid ,Molecular Biology ,RNA, Double-Stranded ,030304 developmental biology ,Coronavirus ,Swine Diseases ,Original Paper ,0303 health sciences ,biology ,030306 microbiology ,Activator (genetics) ,RNA-Binding Proteins ,virus diseases ,Porcine deltacoronavirus ,MDA5 ,Interferon-beta ,General Medicine ,Nucleocapsid Proteins ,RNA binding ,biology.organism_classification ,Protein kinase R ,HEK293 Cells ,DEAD Box Protein 58 ,Interferon Regulatory Factor-3 ,Coronavirus Infections ,Protein Binding ,medicine.drug - Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus that causes watery diarrhea, vomiting and mortality in newborn piglets. Previous studies have suggested that PDCoV infection antagonizes RIG-I-like receptor (RLR)-mediated IFN-β production to evade host innate immune defense, and PDCoV-encoded nonstructural protein nsp5 and accessory protein NS6 are associated with this process. However, whether the structural protein(s) of PDCoV also antagonize IFN-β production remains unclear. In this study, we found that PDCoV nucleocapsid (N) protein, the most abundant viral structural protein, suppressed Sendai virus (SEV)-induced IFN-β production and transcription factor IRF3 activation, but did not block IFN-β production induced by overexpressing RIG-I/MDA5. Furthermore, study revealed that PDCoV N protein interacted with RIG-I and MDA5 in an in vitro overexpression system and evident interactions between N protein and RIG-I could be detected in the context of PDCoV infection, which interfered with the binding of dsRNA and protein activator of protein kinase R (PACT) to RIG-I. Together, our results demonstrate that PDCoV N protein is an IFN antagonist and utilizes diverse strategies to attenuate RIG-I recognition and activation.
- Published
- 2019
- Full Text
- View/download PDF
29. RACK1 attenuates RLR antiviral signaling by targeting VISA-TRAF complexes
- Author
-
Lingzhen Cao, Tian Chen, Liang-Guo Xu, Changsheng Li, Weiying Wang, Hua Rao, Qing Yang, Tao Xie, and Hong-Bing Shu
- Subjects
0301 basic medicine ,Small interfering RNA ,viruses ,Biophysics ,Protein Serine-Threonine Kinases ,Receptors for Activated C Kinase ,Antiviral Agents ,Sendai virus ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,TANK-binding kinase 1 ,Interferon ,medicine ,Humans ,RNA, Small Interfering ,Promoter Regions, Genetic ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,RIG-I ,Chemistry ,Lysine ,Ubiquitination ,Receptor for activated C kinase 1 ,Interferon-beta ,Cell Biology ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,Neoplasm Proteins ,Cell biology ,030104 developmental biology ,Gene Knockdown Techniques ,Multiprotein Complexes ,030220 oncology & carcinogenesis ,DEAD Box Protein 58 ,Signal transduction ,IRF3 ,Signal Transduction ,medicine.drug ,Interferon regulatory factors - Abstract
Virus-induced signaling adaptor (VISA), which mediates the production of type I interferon, is crucial for the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) signaling pathway. Upon viral infection, RIG-I recognizes double-stranded viral RNA and interacts with VISA to mediate antiviral innate immunity. However, the mechanisms underlying RIG/VISA-mediated antiviral regulation remain unclear. In this study, we confirmed that receptor for activated C kinase 1 (RACK1) interacts with VISA and attenuates the RIG/VISA-mediated antiviral innate immune signaling pathway. Overexpression of RACK1 inhibited the interferon-β (IFN-β) promoter; interferon-stimulated response element (ISRE); nuclear factor kappa B (NF-κB) activation; and dimerization of interferon regulatory factor 3 (IRF3) mediated by RIG-I, VISA, and TANK-binding kinase 1 (TBK1). A reduction in RACK1 expression level upon small interfering RNA knockdown increased RIG/VISA-mediated antiviral transduction. Additionally, RACK1 disrupted formation of the VISA-tumor necrosis factor receptor-associated factor 2 (TRAF2), VISA-TRAF3, and VISA-TRAF6 complexes during RIG-I/VISA-mediated signal transduction. Additionally, RACK1 enhanced K48-linked ubiquitination of VISA, attenuated its K63-linked ubiquitination, and decreased VISA-mediated antiviral signal transduction. Together, these results indicate that RACK1 interacts with VISA to repress downstream signaling and downregulates virus-induced IFN-β production in the RIG-I/VISA signaling pathway.
- Published
- 2019
- Full Text
- View/download PDF
30. Generation of two human induced pluripotent stem cell lines from patients with biallelic USH2A variants
- Author
-
Zixi Sun, Tian Zhu, Ruifang Sui, Xuan Zou, Xing Wei, Xiaoxu Han, and Shijing Wu
- Subjects
QH301-705.5 ,Usher syndrome ,Induced Pluripotent Stem Cells ,Biology ,Peripheral blood mononuclear cell ,otorhinolaryngologic diseases ,medicine ,Humans ,Biology (General) ,Induced pluripotent stem cell ,Gene ,Extracellular Matrix Proteins ,Karyotype ,Cell Biology ,General Medicine ,biology.organism_classification ,medicine.disease ,Molecular biology ,eye diseases ,Sendai virus ,Mutation ,Leukocytes, Mononuclear ,Autosomal recessive retinitis pigmentosa ,Usher Syndromes ,Retinitis Pigmentosa ,Developmental Biology - Abstract
Usher syndrome 2A (USH2A) is one of the most common genes associated with Usher syndrome type II (USH2) and nonsyndromic autosomal recessive retinitis pigmentosa (arRP). Here, we describe the generation and characterization of two human induced pluripotent stem cell (hiPSC) lines from a RP patient with compound heterogeneous USH2A variants and a USH2 patient with homozygous USH2A variant. Blood samples were obtained and peripheral blood mononuclear cells (PBMCs) were reprogrammed using the non-integrative Sendai virus to generate the iPSC lines. The established hiPSC lines retained the disease-associated variants and showed normal karyotype, pluripotency and differentiation capacity.
- Published
- 2021
31. SARS-CoV-2 3C-like protease antagonizes interferon-beta production by facilitating the degradation of IRF3
- Author
-
Zhenling Ma, Wenwen Zhang, Menghao Zhang, Lei Wang, Yaru Wu, Wei Liu, Xixi Shi, Yanyan Zhang, and Min Zhang
- Subjects
SARS-CoV-2 3CL ,medicine.medical_treatment ,viruses ,Immunology ,Biology ,Response Elements ,Biochemistry ,Sendai virus ,Article ,Immune system ,medicine ,Immunology and Allergy ,Humans ,RNA, Messenger ,Receptors, Immunologic ,DEAD Box Protein 58 ,skin and connective tissue diseases ,Promoter Regions, Genetic ,Molecular Biology ,Coronavirus 3C Proteases ,Innate immune system ,Protease ,fungi ,NF-kappa B ,virus diseases ,Hematology ,Transfection ,Interferon-beta ,biochemical phenomena, metabolism, and nutrition ,IRF3 ,biology.organism_classification ,Virology ,HEK293 Cells ,Gene Expression Regulation ,Interferon-beta production ,Proteolysis ,Innate immune ,Interferon Regulatory Factor-3 ,RLR signalling pathway ,Signal Transduction ,IFNs - Abstract
The prevalence of SARS-CoV-2 is a great threat to global public health. However, the relationship between the viral pathogen SARS-CoV-2 and host innate immunity has not yet been well studied. The genome of SARS-CoV-2 encodes a viral protease called 3C-like protease. This protease is responsible for cleaving viral polyproteins during replication. In this investigation, 293T cells were transfected with SARS-CoV-2 3CL and then infected with Sendai virus (SeV) to induce the RIG-I like receptor (RLR)-based immune pathway. q-PCR, luciferase reporter assays, and western blotting were used for experimental analyses. We found that SARS-CoV-2 3CL significantly downregulated IFN-β mRNA levels. Upon SeV infection, SARS-CoV-2 3CL inhibited the nuclear translocation of IRF3 and p65 and promoted the degradation of IRF3. This effect of SARS-CoV-2 3CL on type I IFN in the RLR immune pathway opens up novel ideas for future research on SARS-CoV-2.
- Published
- 2021
32. Direct reprogramming with Sendai virus vectors repaired infarct hearts at the chronic stage
- Author
-
Taketaro Sadahiro, Ryo Fujita, Hiroaki Mizukami, Mari Isomi, Masaki Ieda, Keiichi Fukuda, Tatsuya Akiyama, Tsugumine Shu, Yu Yamada, and Yuto Abe
- Subjects
0301 basic medicine ,Cardiac function curve ,Male ,Genetic Vectors ,Biophysics ,Myocardial Infarction ,Biochemistry ,Sendai virus ,Collagen Type I ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,medicine ,Animals ,MEF2C ,Myocytes, Cardiac ,Myocardial infarction ,Ventricular remodeling ,Molecular Biology ,biology ,business.industry ,Myocardium ,Cell Biology ,Fibroblasts ,medicine.disease ,biology.organism_classification ,Cellular Reprogramming ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,Heart failure ,Chronic Disease ,cardiovascular system ,Cancer research ,business ,Reprogramming ,Transcription Factors - Abstract
Adult hearts have limited regenerative capacity. Hence, after acute myocardial infarction (MI), dead myocardial tissues are digested by immune cells and replaced by fibrosis, leading to ventricular remodeling and heart failure at the chronic stage. Direct reprogramming of the cardiac fibroblasts (CFs) into induced cardiomyocytes (iCMs) with cardiac transcription factors, including Gata4, Mef2c, and Tbx5 (GMT), may have significant potential for cardiac repair. Sendai virus (SeV) vectors expressing GMT have been reported to reprogram the mouse cardiac fibroblasts into iCMs without any risk of insertional mutagenesis. In vivo reprogramming improved the cardiac function after acute MI in immunodeficient mice. However, it is unknown whether the newly generated iCMs could exist in infarct hearts for a prolonged period and SeV-GMT can improve cardiac function after MI at the chronic stage in immunocompetent mice. Here, we show that SeV vectors efficiently infect CFs in vivo and reprogram them into iCMs, which existed for at least four weeks after MI, in fibroblast-linage tracing mice. Moreover, SeV-GMT improved cardiac function and reduced fibrosis and collagen I expression at 12 weeks after MI in immunocompetent mice. Thus, direct cardiac reprogramming with SeV vectors could be a promising therapy for MI.
- Published
- 2021
33. JMJD6 negatively regulates cytosolic RNA induced antiviral signaling by recruiting RNF5 to promote activated IRF3 K48 ubiquitination
- Author
-
Yueyue Duan, Junwu Ma, Qi Wang, Zhang Keshan, Zixiang Zhu, Xiangtao Liu, Zhang Wei, Zheng Haixue, Fan Yang, Weijun Cao, and Yang Yang
- Subjects
Proteomics ,Jumonji Domain-Containing Histone Demethylases ,viruses ,Biochemistry ,Mice ,0302 clinical medicine ,Interferon ,Biology (General) ,Post-Translational Modification ,Phosphorylation ,0303 health sciences ,biology ,Small interfering RNA ,Sendai virus ,Cell biology ,Precipitation Techniques ,Enzymes ,Nucleic acids ,DNA-Binding Proteins ,Vesicular stomatitis virus ,Oxidoreductases ,Luciferase ,medicine.drug ,Research Article ,Signal Transduction ,QH301-705.5 ,Ubiquitin-Protein Ligases ,Immunology ,DNA construction ,Research and Analysis Methods ,Transfection ,Microbiology ,Antiviral Agents ,Virus ,03 medical and health sciences ,Virology ,medicine ,Genetics ,Immunoprecipitation ,Animals ,Humans ,Molecular Biology Techniques ,Non-coding RNA ,Molecular Biology ,030304 developmental biology ,Mitochondrial antiviral-signaling protein ,Ubiquitination ,RNA ,Biology and Life Sciences ,Proteins ,Membrane Proteins ,RNA virus ,RC581-607 ,biology.organism_classification ,Gene regulation ,Plasmid Construction ,Enzymology ,Parasitology ,Interferon Regulatory Factor-3 ,Gene expression ,Immunologic diseases. Allergy ,IRF3 ,030217 neurology & neurosurgery ,Viral Transmission and Infection - Abstract
The negative regulation of antiviral immune responses is essential for the host to maintain homeostasis. Jumonji domain-containing protein 6 (JMJD6) was previously identified with a number of functions during RNA virus infection. Upon viral RNA recognition, retinoic acid-inducible gene-I-like receptors (RLRs) physically interact with the mitochondrial antiviral signaling protein (MAVS) and activate TANK-binding kinase 1 (TBK1) to induce type-I interferon (IFN-I) production. Here, JMJD6 was demonstrated to reduce type-I interferon (IFN-I) production in response to cytosolic poly (I:C) and RNA virus infections, including Sendai virus (SeV) and Vesicular stomatitis virus (VSV). Genetic inactivation of JMJD6 enhanced IFN-I production and impaired viral replication. Our unbiased proteomic screen demonstrated JMJD6 contributes to IRF3 K48 ubiquitination degradation in an RNF5-dependent manner. Mice with gene deletion of JMJD6 through piggyBac transposon-mediated gene transfer showed increased VSV-triggered IFN-I production and reduced susceptibility to the virus. These findings classify JMJD6 as a negative regulator of the host’s innate immune responses to cytosolic viral RNA., Author summary RLRs-mediated signaling needs to be terminated in order to prevent persistent immune responses and adverse effects to the host once the virus has been cleared. In this study, we provide rigorous evidence that JMJD6 negatively regulates RLRs-mediated innate immune responses. We found that JMJD6 recruits RNF5 to induce the K48-linked polyubiquitination and proteasomal degradation of activated IRF3. Genetic inactivation of JMJD6 in cells increases IFN-I production to suppress viral infection. Consistently, in vivo studies show that, compared with WT mice, JMJD6-deficient mice are more resistant to VSV infection with more IFN-I production and reduced viral load in livers. Our findings reveal a novel mechanism to downregulate innate immune responses mediated by RNA viral infection, which allows the host to prevent undue immune responses and sustain homeostasis.
- Published
- 2021
34. Porcine deltacoronavirus nsp10 antagonizes interferon-β production independently of its zinc finger domains
- Author
-
Puxian Fang, Yanrong Zhou, Yingying Hong, Jie Ren, Sijin Xia, Liurong Fang, Shaobo Xiao, and Jiansong Zhang
- Subjects
Swine ,Viral Nonstructural Proteins ,medicine.disease_cause ,Sendai virus ,Cell Line ,03 medical and health sciences ,Interferon ,Virology ,medicine ,Animals ,Humans ,Transcription factor ,030304 developmental biology ,Coronavirus ,Zinc finger ,0303 health sciences ,biology ,030302 biochemistry & molecular biology ,Transcription Factor RelA ,Zinc Fingers ,Interferon-beta ,biology.organism_classification ,Molecular biology ,Host-Pathogen Interactions ,Mutation ,Phosphorylation ,Ectopic expression ,Interferon Regulatory Factor-3 ,IRF3 ,Deltacoronavirus ,medicine.drug ,Signal Transduction - Abstract
Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that causes serious vomiting and diarrhea in piglets. Previous work demonstrated that PDCoV infection inhibits type I interferon (IFN) production. Here, we found that ectopic expression of PDCoV nsp10 significantly inhibited Sendai virus (SeV)-induced IFN-β production by impairing the phosphorylation and nuclear translocation of two transcription factors, IRF3 and NF-κB p65 subunit. Interestingly, experiments with truncated mutants and site-directed mutagenesis revealed that PDCoV nsp10 mutants with missing or destroyed zinc fingers (ZFs) domains also impeded SeV-induced IFN-β production, suggesting that nsp10 does not require its ZF domains to antagonize IFN-β production. Further work found that co-expression of nsp10 with nsp14 or nsp16, two replicative enzymes, significantly enhanced the inhibitory effects of nsp10 on IFN-β. Taken together, our results demonstrate that PDCoV nsp10 antagonizes IFN via a ZF-independent mechanism and has a synergistic effect with nsp14 and nsp16 on inhibiting IFN-β production.
- Published
- 2021
35. Generation of an induced pluripotent stem cell line from a Bartter syndrome patient with the homozygote mutation p.A244D (c.731CA) in SLC12A1 gene
- Author
-
Xing Rong, Xian Shen, Xiaoling Guo, Dexuan Wang, Chao Li, Yinjuan Ding, Weiping Ji, Congde Chen, Maoping Chu, Huihui Chen, and Xiaoou Shan
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Gene mutation ,Bartter syndrome ,medicine.disease_cause ,Sendai virus ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Induced pluripotent stem cell ,Gene ,lcsh:QH301-705.5 ,Solute Carrier Family 12, Member 1 ,Mutation ,biology ,Homozygote ,Bartter Syndrome ,Karyotype ,Cell Differentiation ,Cell Biology ,General Medicine ,biology.organism_classification ,medicine.disease ,Molecular biology ,030104 developmental biology ,lcsh:Biology (General) ,Child, Preschool ,Female ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Bartter Syndrome (BS) is a group of rare inherited autosome-recessive disease, which can be caused by the gene mutations of sodium-potassium-chloride cotransporter gene (SLC12A1). Here, the urine cells (UCs) derived from a 4-year-old female BS patient with the homozygote SLC12A1 gene mutation p.A244D (c.731C>A) were reprogramming into induced pluripotent stem cells (iPSCs) named WMUi019-A using a commercial Sendai virus reprogramming kit. The pluripotent stem cell markers like OCT4 and SSEA4 can be positively expressed in this iPSC line, which can also be induced to differentiate into three germ layers in vitro and maintain a stable karyotype (46, XY).
- Published
- 2021
36. S67 Low levels of lentivirus-mediated CFTR gene transfer are sufficient to generate ion transport correction in air-liquid interface cultures from cystic fibrosis patients
- Author
-
S Kreuz, Stephen C. Hyde, A Moiseenko, N A Nafchi, A Sergijenko, K M Pineault, T Gamlen, Mario Chan, Ewfw Alton, Uta Griesenbach, and Gill
- Subjects
Forskolin ,biology ,business.industry ,biology.organism_classification ,Molecular biology ,Sendai virus ,Amiloride ,Viral vector ,chemistry.chemical_compound ,chemistry ,DIDS ,In vivo ,Medicine ,business ,Ex vivo ,Ion transporter ,medicine.drug - Abstract
Introduction We have developed a lentiviral vector pseudotyped with the F and HN proteins from Sendai virus (rSIV.F/HN) for cystic fibrosis (CF) gene therapy and are now progressing towards a first-in-man clinical trial. Here we assessed the transduction efficiency of rSIV.F/HN expressing EGFP in human bronchial epithelial cells from healthy control (HC) and CF donors grown in air-liquid interface culture (ALI). We also assessed the degree of correction of ion transport by rSIV.F/HN-CFTR in this model. Methods Fully differentiated ALIs (MucilAir, Epithelix) were transduced with rSIV.F/HN-EGFP or Sendai virus (SeV)-GFP and GFP expression was quantified at multiple time points using fluorescence microscopy or flow cytometry. The ion transport in HC, CF, and CF ALIs transduced with rSIV.F/HN-CFTR was measured at 7 days in Ussing chambers (stepwise protocol: chloride buffer as baseline, 100 μM amiloride, 100 μM DIDS, low chloride, 10 μM forskolin/100 μM IBMX, 30 μM ΔCFTR inhibitor-172). Results Transduction efficiency was generally In Ussing chambers, there was no difference in baseline short circuit current between HC and CF ALIs, while forskolin/IBMX-mediated chloride secretion was significantly higher in HC samples compared to CF (HC: 8.9±1.4 µA/cm2, CF: 1.16±0.33 µA/cm2, n=14–17/group). We then assessed whether transduction of CF ALIs with rSIV.F/HN-CFTR was able to correct the chloride transport defect. Chloride transport increased significantly (p Conclusion These data suggest that ex vivo transduction efficiency of differentiated human ALIs is low and may not reflect the in vivo performance of gene transfer agents. However, even at this low transduction efficiency, functional correction of ~40% of chloride transport was achieved in CF patient-derived ALI cultures following transduction with rSIV.F/HN-CFTR.
- Published
- 2021
- Full Text
- View/download PDF
37. A human surfactant B deficiency air-liquid interface cell culture model suitable for gene therapy applications
- Author
-
Altar M. Munis, Stephen C. Hyde, and Deborah R. Gill
- Subjects
0301 basic medicine ,Surfactant B deficiency ,lcsh:QH426-470 ,Alveolar Epithelium ,Genetic enhancement ,lentiviral vectors ,Viral vector ,Alveolar cells ,03 medical and health sciences ,0302 clinical medicine ,ALI culture model ,lentivirus ,Parenchyma ,Genetics ,medicine ,interstitial lung diseases ,rSIV.F/HN ,lcsh:QH573-671 ,Molecular Biology ,chemistry.chemical_classification ,biology ,lcsh:Cytology ,biology.organism_classification ,gene therapy ,Phenotype ,Sendai virus ,Cell biology ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,SPB ,Molecular Medicine ,Original Article ,Glycoprotein ,ATII cells - Abstract
Surfactant protein B (SPB) deficiency is a severe monogenic interstitial lung disorder that leads to loss of life in infants as a result of alveolar collapse and respiratory distress syndrome. The development and assessment of curative therapies for the deficiency are limited by the general lack of well-characterized and physiologically relevant in vitro models of human lung parenchyma. Here, we describe a new human surfactant air-liquid interface (SALI) culture model based on H441 cells, which successfully recapitulates the key characteristics of human alveolar cells in primary culture as evidenced by RNA and protein expression of alveolar cell markers. SALI cultures were able to develop stratified cellular layers with functional barrier properties that are stable for at least 28 days after air-lift. A SFTPB knockout model of SPB deficiency was generated via gene editing of SALI cultures. The SFTPB-edited SALI cultures lost expression of SPB completely and showed weaker functional barrier properties. We were able to correct this phenotype via delivery of a lentiviral vector pseudotyped with Sendai virus glycoproteins F/HN expressing SPB. We believe that SALI cultures can serve as an important in vitro research tool to study human alveolar epithelium, especially for the development of advanced therapy medicinal products targeting monogenic disorders., Graphical Abstract, Munis et al. describe a new human pulmonary air-liquid interface (ALI) cell culture model that recapitulates alveolar cell markers, is physiologically relevant, and facilitates high-throughput assessment of therapeutic interventions. They demonstrate that surfactant protein B deficiency, modeled via SFTPB gene knockout, can be reversed by rSIV.F/HN-mediated gene therapy.
- Published
- 2021
38. TRK-Fused Gene (TFG), a protein involved in protein secretion pathways, is an essential component of the antiviral innate immune response
- Author
-
Anne-Claude Gingras, Marc J. Servant, Kashif Aziz Khan, Alexandre Marineau, Étienne Durette, Mariana Acevedo, and Priscilla Doyon
- Subjects
TRAF3 ,Small interfering RNA ,Cultured tumor cells ,Biochemistry ,Medicine and Health Sciences ,Post-Translational Modification ,Phosphorylation ,Biology (General) ,Immune Response ,COPII ,0303 health sciences ,Secretory Pathway ,biology ,030302 biochemistry & molecular biology ,Sendai virus ,Precipitation Techniques ,3. Good health ,Ubiquitin ligase ,Cell biology ,Nucleic acids ,Immunoblot Analysis ,Viperin ,Interferon Type I ,Cell lines ,Signal transduction ,Biological cultures ,Research Article ,Signal Transduction ,QH301-705.5 ,Immunology ,Molecular Probe Techniques ,Protein Serine-Threonine Kinases ,Transfection ,Antiviral Agents ,Microbiology ,03 medical and health sciences ,Rhabdoviridae Infections ,Virology ,Genetics ,Immunoprecipitation ,Humans ,HeLa cells ,Non-coding RNA ,Molecular Biology Techniques ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Endoplasmic reticulum membrane ,TNF Receptor-Associated Factor 3 ,Biology and Life Sciences ,Proteins ,Vesiculovirus ,RC581-607 ,Cell cultures ,biology.organism_classification ,Immunity, Innate ,Gene regulation ,Research and analysis methods ,Antiviral Immune Response ,biology.protein ,RNA ,Parasitology ,Gene expression ,Immunologic diseases. Allergy ,Viral Transmission and Infection - Abstract
Antiviral innate immune response to RNA virus infection is supported by Pattern-Recognition Receptors (PRR) including RIG-I-Like Receptors (RLR), which lead to type I interferons (IFNs) and IFN-stimulated genes (ISG) production. Upon sensing of viral RNA, the E3 ubiquitin ligase TNF Receptor-Associated Factor-3 (TRAF3) is recruited along with its substrate TANK-Binding Kinase (TBK1), to MAVS-containing subcellular compartments, including mitochondria, peroxisomes, and the mitochondria-associated endoplasmic reticulum membrane (MAM). However, the regulation of such events remains largely unresolved. Here, we identify TRK-Fused Gene (TFG), a protein involved in the transport of newly synthesized proteins to the endomembrane system via the Coat Protein complex II (COPII) transport vesicles, as a new TRAF3-interacting protein allowing the efficient recruitment of TRAF3 to MAVS and TBK1 following Sendai virus (SeV) infection. Using siRNA and shRNA approaches, we show that TFG is required for virus-induced TBK1 activation resulting in C-terminal IRF3 phosphorylation and dimerization. We further show that the ability of the TRAF3-TFG complex to engage mTOR following SeV infection allows TBK1 to phosphorylate mTOR on serine 2159, a post-translational modification shown to promote mTORC1 signaling. We demonstrate that the activation of mTORC1 signaling during SeV infection plays a positive role in the expression of Viperin, IRF7 and IFN-induced proteins with tetratricopeptide repeats (IFITs) proteins, and that depleting TFG resulted in a compromised antiviral state. Our study, therefore, identifies TFG as an essential component of the RLR-dependent type I IFN antiviral response., Author summary Antiviral innate immune response is the first line of defence against the invading viruses through type I interferon (IFN) signaling. However, viruses have devised ways to target signaling molecules for aberrant IFN response and worsen the disease outcome. As such, deciphering the roles of new regulators of innate immunity could transform the antiviral treatment paradigm by introducing novel panviral therapeutics designed to reinforce antiviral host responses. This could be of great use in fighting recent outbreaks of severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome MERS-CoV, and the more recent SARS-CoV-2 causing the COVID-19 pandemic. However, aberrant activation of such pathways can lead to detrimental consequences, including autoimmune diseases. Regulation of type I IFN responses is thus of paramount importance. To prevent an uncontrolled response, signaling events happen in discrete subcellular compartments, therefore, distinguishing sites involved in recognition of pathogens and those permitting downstream signaling. Here, we show TFG as a new regulator of type I IFN response allowing the efficient organization of signaling molecules. TFG, thus, further substantiates the importance of the protein trafficking machinery in the regulation of optimal antiviral responses. Our findings have implications for both antiviral immunity and autoimmune diseases.
- Published
- 2021
39. Comparison of Temporal Transcriptomic Profiles from Immature Lungs of Two Rat Strains Reveals a Viral Response Signature Associated with Chronic Lung Dysfunction.
- Author
-
Hines, Elizabeth A., Szakaly, Renee J., Leng, Ning, Webster, Anais T., Verheyden, Jamie M., Lashua, Amber J., Kendziorski, Christina, Rosenthal, Louis A., Gern, James E., Sorkness, Ronald L., Sun, Xin, and Jr.Lemanske, Robert F.
- Subjects
- *
ASTHMA in children , *VIRUS diseases , *SENDAI virus , *IMMUNE response , *GENE expression , *ANTIBODY-dependent cell cytotoxicity - Abstract
Early life respiratory viral infections and atopic characteristics are significant risk factors for the development of childhood asthma. It is hypothesized that repeated respiratory viral infections might induce structural remodeling by interfering with the normal process of lung maturation; however, the specific molecular processes that underlie these pathological changes are not understood. To investigate the molecular basis for these changes, we used an established Sendai virus infection model in weanling rats to compare the post-infection transcriptomes of an atopic asthma susceptible strain, Brown Norway, and a non-atopic asthma resistant strain, Fischer 344. Specific to this weanling infection model and not described in adult infection models, Sendai virus in the susceptible, but not the resistant strain, results in morphological abnormalities in distal airways that persist into adulthood. Gene expression data from infected and control lungs across five time points indicated that specific features of the immune response following viral infection were heightened and prolonged in lungs from Brown Norway rats compared with Fischer 344 rats. These features included an increase in macrophage cell number and related gene expression, which then transitioned to an increase in mast cell number and related gene expression. In contrast, infected Fischer F344 lungs exhibited more efficient restoration of the airway epithelial morphology, with transient appearance of basal cell pods near distal airways. Together, these findings indicate that the pronounced macrophage and mast cell responses and abnormal re-epithelialization precede the structural defects that developed and persisted in Brown Norway, but not Fischer 344 lungs. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
40. Generation of an iPSC line (AKOSi006-A) from fibroblasts of an NPC1 patient, carrying the homozygous mutation p.I1061T (c.3182 T > C) and a control iPSC line (AKOSi007-A) using a non-integrating Sendai virus system
- Author
-
Moritz J. Frech, Christin Völkner, Andreas Hermann, Jörn Bullerdiek, Gudrun Knuebel, Maik Liedtke, Katharina Huth, Janine Petters, Jan Lukas, and Hugo Murua Escobar
- Subjects
0301 basic medicine ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Lipid storage disorder ,Induced Pluripotent Stem Cells ,genetics [Mutation] ,medicine.disease_cause ,Sendai virus ,03 medical and health sciences ,Kruppel-Like Factor 4 ,0302 clinical medicine ,KLF4 protein, human ,Niemann-Pick C1 Protein ,ddc:570 ,hemic and lymphatic diseases ,medicine ,Humans ,NPC1 protein, human ,lcsh:QH301-705.5 ,Mutation ,biology ,Intracellular Signaling Peptides and Proteins ,nutritional and metabolic diseases ,Niemann-Pick Disease, Type C ,Cell Biology ,General Medicine ,Fibroblasts ,biology.organism_classification ,medicine.disease ,Sphingolipid ,Molecular biology ,030104 developmental biology ,lcsh:Biology (General) ,KLF4 ,Cell culture ,genetics [Sendai virus] ,lipids (amino acids, peptides, and proteins) ,NPC1 ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Niemann-Pick disease type C1 (NPC1) is a rare inherited lipid storage disorder caused by mutations in the NPC1 gene. Mutations lead to impaired lipid trafficking and subsequently to accumulation of cholesterol and sphingolipids. NPC1-patients present variable multisystemic symptoms, including neurological deficits. Here, we describe the generation of human iPSC lines obtained from fibroblasts of a male individual, carrying the homozygous mutation p.I1061T, and an unrelated and healthy male individual. A non-integrating Sendai virus system, containing KLF4, OCT3/4, SOX2 and C-MYC, was used for reprogramming. These cell lines provide a valuable resource for studying the pathophysiology of multisystemic NPC1-disease.
- Published
- 2020
- Full Text
- View/download PDF
41. Generation of a human induced pluripotent stem cell line (CMCi002-A) from a patient with Gitelman’s syndrome
- Author
-
Yoo-Jin Shin, Eun Jeong Ko, Sun Woo Lim, Chul Woo Yang, Sheng Cui, Jae Young Lee, Byung Ha Chung, and Kang In Lee
- Subjects
Adult ,Male ,Induced Pluripotent Stem Cells ,Germ layer ,medicine.disease_cause ,Peripheral blood mononuclear cell ,Cell Line ,medicine ,Humans ,Solute Carrier Family 12, Member 3 ,Induced pluripotent stem cell ,Gene ,lcsh:QH301-705.5 ,Mutation ,biology ,Karyotype ,Human induced pluripotent stem cells ,Cell Biology ,General Medicine ,biology.organism_classification ,Embryonic stem cell ,Molecular biology ,SLC12A3 gene ,Sendai virus ,lcsh:Biology (General) ,Leukocytes, Mononuclear ,Gitelman’s syndrome ,Gitelman Syndrome ,Developmental Biology - Abstract
We established a human induced pluripotent stem cells (hiPSC) line (CMCi002-A) from peripheral blood mononuclear cells (PBMCs) of 29-year-old male with Gitelman's syndrome (GIT) caused by the mutation of solute carrier family 12 member 3 (SLC12A3) gene using Sendai virus. The GIT-hiPSCs showed a typical human embryonic stem cell like morphology and expressed all pluripotency-associated markers, exhibited normal karyotype and were capable of differentiating into cells representative of three germ layers.
- Published
- 2020
42. SARS-CoV-2 evades immune detection in alveolar macrophages
- Author
-
Line S. Reinert, David Olagnier, Rune Hartmann, Jan J. Enghild, Jacob Thyrsted, Louise Dalskov, Michelle Møhlenberg, Hans Jürgen Hoffmann, Ebbe Toftgaard Poulsen, Søren Helbo Skaarup, Christian K. Holm, Julia Blay-Cadanet, and Birgitte Holst Folkersen
- Subjects
viruses ,medicine.disease_cause ,Biochemistry ,Antiviral Agents ,Virus ,SARS‐CoV‐2 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Interferon ,COVID‐19 ,Report ,Macrophages, Alveolar ,Genetics ,medicine ,Influenza A virus ,Humans ,Respiratory system ,Molecular Biology ,Lung ,Pandemics ,Cells, Cultured ,030304 developmental biology ,Immune Evasion ,0303 health sciences ,biology ,business.industry ,SARS-CoV-2 ,interferon lambda ,COVID-19 ,Epithelial Cells ,alveolar macrophages ,interferon ,biology.organism_classification ,RNA Biology ,Sendai virus ,medicine.anatomical_structure ,Immunology ,Interferon Type I ,Cytokines ,business ,Development & Differentiation ,030217 neurology & neurosurgery ,medicine.drug ,Respiratory tract ,Reports - Abstract
Respiratory infections, like the current COVID‐19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue‐resident macrophages located within the lung. They play a key role in the early phases of an immune response to respiratory viruses. AMs are likely the first immune cells to encounter SARS‐CoV‐2 during an infection, and their reaction to the virus will have a profound impact on the outcome of the infection. Interferons (IFNs) are antiviral cytokines and among the first cytokines produced upon viral infection. In this study, AMs from non‐infectious donors are challenged with SARS‐CoV‐2. We demonstrate that challenged AMs are incapable of sensing SARS‐CoV‐2 and of producing an IFN response in contrast to other respiratory viruses, like influenza A virus and Sendai virus, which trigger a robust IFN response. The absence of IFN production in AMs upon challenge with SARS‐CoV‐2 could explain the initial asymptotic phase observed during COVID‐19 and argues against AMs being the sources of pro‐inflammatory cytokines later during infection., Alveolar macrophages produce interferons and activate interferon stimulated genes when challenged with influenza A virus or Sendai virus, but not when challenged with SARS‐CoV‐2, suggesting that its genomic RNA is undetectable for innate immune sensors.
- Published
- 2020
- Full Text
- View/download PDF
43. Generation of the iPSC line IISHDOi007-A from peripheral blood mononuclear cells from a patient with McArdle disease harbouring the mutation c.2392 T > C; p.Trp798Arg
- Author
-
Ana Moreno-Izquierdo, Victoria Cerrada, Sofía Alvarez-Galeano, Alejandro Lucia, M. Esther Gallardo, Marta García-López, María Rabasa Pérez, and Joaquín Arenas
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Biology ,medicine.disease_cause ,Peripheral blood mononuclear cell ,Cell Line ,03 medical and health sciences ,Kruppel-Like Factor 4 ,0302 clinical medicine ,medicine ,Humans ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Mutation ,Cell Biology ,General Medicine ,medicine.disease ,biology.organism_classification ,Molecular biology ,Sendai virus ,030104 developmental biology ,lcsh:Biology (General) ,KLF4 ,Cell culture ,embryonic structures ,Leukocytes, Mononuclear ,Glycogen Storage Disease Type V ,Reprogramming ,030217 neurology & neurosurgery ,Glycogen storage disease type V ,Developmental Biology - Abstract
Peripheral blood mononuclear cells (PBMCs) from a McArdle patient carrying a homozygous mutation in the PYGM gene: c.2392 T > C; p.Trp798Arg were used for the generation of the human iPSC line, IISHDOi007-A. For the delivery of the reprogramming factors Oct3/4, Sox2, Klf4, and c-Myc, a non-integrative methodology that implies the use of Sendai virus has been applied.
- Published
- 2020
44. Generation of four iPSC lines from Neurofibromatosis Type 1 patients
- Author
-
Wei Shern Lee, Jonathan M. Payne, Kristina M Haebich, Kathryn N. North, Paul J. Lockhart, and Kiymet Bozaoglu
- Subjects
0301 basic medicine ,Homeobox protein NANOG ,Neurofibromatosis 1 ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Biology ,Immunofluorescence ,Sendai virus ,03 medical and health sciences ,Kruppel-Like Factor 4 ,0302 clinical medicine ,SOX2 ,medicine ,Humans ,Neurofibromatosis ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,medicine.diagnostic_test ,Cell Differentiation ,Cell Biology ,General Medicine ,medicine.disease ,Cellular Reprogramming ,Molecular biology ,030104 developmental biology ,lcsh:Biology (General) ,KLF4 ,embryonic structures ,Leukocytes, Mononuclear ,biological phenomena, cell phenomena, and immunity ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
We describe the generation and characterisation of four human induced pluripotent stem cell (iPSC) lines from peripheral blood mononuclear cells (PBMC) from individuals with neurofibromatosis type (NF1). PBMC reprogramming was performed using a non-integrative Sendai virus containing the reprogramming factors OCT4, SOX2, MYC and KLF4. All iPSC lines exhibited a normal karyotype, and pluripotency was validated by flow cytometry (EPCAM, TRA-1-81, SSEA1 and CD9) and immunofluorescence (OCT4 and Nanog). Differentiation of the cells into the three embryonic germ layers was confirmed using immunofluorescence. These iPSC lines are a valuable pre-clinical resource to study the molecular mechanisms underlying NF1.
- Published
- 2020
45. Impaired HVJ-stimulated Interferon producing capacity in MPO-ANCA-associated vasculitis with rapidly progressive glomerulonephritis lead to susceptibility to infection
- Author
-
Kazuo Suzuki, Toshiko Ito-Ihara, Katusmi Yagi, Kazuko Uno, Yuko Yasuda, Tomomi Endo, and Eri Muso
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,T cell ,Immunology ,Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis ,Plasmacytoid dendritic cell ,Sendai virus ,Biochemistry ,Antibodies, Antineutrophil Cytoplasmic ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,medicine ,Rapidly progressive glomerulonephritis ,Humans ,Immunology and Allergy ,Molecular Biology ,Whole blood ,Aged ,Aged, 80 and over ,Kidney ,business.industry ,Interferon-alpha ,Dendritic Cells ,Hematology ,Middle Aged ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Disease Susceptibility ,Calprotectin ,business ,medicine.drug ,Systemic vasculitis - Abstract
ANCA-associated RPGN leads to renal failure through systemic vasculitis and diffuse crescentic glomerulonephritis. MPO-ANCA-RPGN patients are highly susceptible to infections. Our aim in this study was to uncover reasons why these patients were susceptible to infections. We analyzed various aspects of type I interferon system including HVJ-stimulated IFN-α producing capacity and plasmacytoid dendritic cell (pDC) number in whole blood in MPO-ANCA-RPGN patients. Compared with healthy subjects, MPO-ANCA-RPGN patients showed impaired HVJ-stimulated IFN-α producing capacity and lower pDC number with or without glucocorticoid treatment. Immuno-histological staining of MPO-ANCA-RPGN kidney samples revealed a few but apparent pDC in T cell infiltrating regions even in patients with low pDC number in their peripheral blood. Patients' low HVJ-stimulated IFN-α producing capacity and pDC numbers persisted even after patients underwent several years of treatment. Former infection was determined using patients' serum BPI, Lamp-2 and Calprotectin, since they are reflective of a history of infection. These markers were higher in MPO-ANCA-RPGN patients than in healthy subjects. These results indicate that impaired HVJ-stimulated IFN-α production as well as dysfunction of the IFN system might have resulted from a previous bout of infection and can be partially implicated in patients' long-term susceptibility and vulnerability to infection.
- Published
- 2020
- Full Text
- View/download PDF
46. Generation of an urine-derived induced pluripotent stem cell line from a 5-year old X-linked Alport syndrome (X-LAS) patient
- Author
-
Congde Chen, Yinjuan Ding, Zhao Han, Chao Niu, Xiaoling Guo, Maoping Chu, Zhanguo Chen, Weiping Ji, and Hongfei Tong
- Subjects
0301 basic medicine ,Male ,Induced Pluripotent Stem Cells ,Nephritis, Hereditary ,Gene mutation ,Biology ,medicine.disease_cause ,urologic and male genital diseases ,Sendai virus ,03 medical and health sciences ,Kruppel-Like Factor 4 ,0302 clinical medicine ,SOX2 ,medicine ,Humans ,Alport syndrome ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Mutation ,Cell Differentiation ,Cell Biology ,General Medicine ,medicine.disease ,biology.organism_classification ,Molecular biology ,030104 developmental biology ,lcsh:Biology (General) ,KLF4 ,Child, Preschool ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The gene mutations of the collagen type IV alpha 5 chain (COL4A5) can lead to the inherited haematuria to end-stage renal disease X-linked Alport syndrome (X-LAS). The urine cells of a 5-year-old male X-LAS patient carrying a hemizygous COL4A5 gene mutation p.G1433V (c.4298G>T) were reprogrammed to induced pluripotent stem cells (iPSCs) with Sendai virus reprogramming kit containing OCT4, SOX2, c-MYC, and KLF4 Yamanaka factors. The generated iPSC line WMUi015-A stably expressed pluripotent markers, maintained a normal karyotype (46, XY), and had differentiation potential into three germ layers in vitro.
- Published
- 2020
47. Characterization of human induced pluripotent stem cells line JLUEYEi002-A from a 48 year old healthy male
- Author
-
Lidi Liu, Hong Wu, Yan Cheng, Zhenshan Lv, Shaokun Zhang, and Weiquan Gong
- Subjects
0301 basic medicine ,Male ,QH301-705.5 ,Induced Pluripotent Stem Cells ,Karyotype ,Embryoid body ,Germ layer ,Biology ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Humans ,Biology (General) ,Induced pluripotent stem cell ,Cell Differentiation ,Cell Biology ,General Medicine ,Middle Aged ,biology.organism_classification ,Cellular Reprogramming ,Molecular biology ,In vitro ,Sendai virus ,030104 developmental biology ,Cell culture ,Leukocytes, Mononuclear ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Peripheral blood was extracted from a 48-year old healthy male donor. Induced pluripotent stem cells (iPSC) were reprogrammed by sendai virus encoding Klf-4, c-Myc, Oct-4, and Sox-2. The iPSC line showed pluripotency, which was verified by immunofluorescence staining. The iPSC line showed normal karyotype, and could form embryoid bodies in vitro and differentiate into the 3 germ layers in vivo. This cell line can be served as healthy control for studying inherited disease.
- Published
- 2020
48. Generation and characterization of human induced pluripotent stem cells line JLUEYEi001-A from a 45 year old female with Stickler syndrome
- Author
-
Shaokun Zhang, Weiquan Gong, Zhenshan Lv, Bo Yang, Lidi Liu, and Hong Wu
- Subjects
0301 basic medicine ,QH301-705.5 ,Hearing Loss, Sensorineural ,Induced Pluripotent Stem Cells ,Germ layer ,Embryoid body ,medicine.disease_cause ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Humans ,Stickler syndrome ,Biology (General) ,Induced pluripotent stem cell ,Connective Tissue Diseases ,Mutation ,biology ,Arthritis ,Retinal Detachment ,Karyotype ,Cell Differentiation ,Cell Biology ,General Medicine ,Middle Aged ,biology.organism_classification ,medicine.disease ,Cellular Reprogramming ,Molecular biology ,Sendai virus ,030104 developmental biology ,Female ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Peripheral blood was extracted from a 45-year old female patient clinically diagnosed with Stickler syndrome harboring a heterozygous splicing mutation in COL2A1 (NM_033150, IVS22-1C>T). Induced pluripotent stem cells (iPSC) were reprogrammed by sendai virus encoding Klf-4, c-Myc, Oct-4, and Sox-2. The iPSC line showed pluripotency, which was verified by immunofluorescence staining. The iPSC line showed normal karyotype, and could form embryoid bodies in vitro and differentiate into the 3 germ layers in vivo. This in vitro cellular model can be used to study the pathogenesis underlying Stickler syndrome.
- Published
- 2020
49. Reprogramming of a human induced pluripotent stem cell line from a Marfan syndrome patient harboring a heterozygous mutation of c.2939G A in FBN1 gene
- Author
-
Xinxuan Gao, Xue Sun, Liqiang Sun, Zhiping Qin, and Hang Su
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Marfan syndrome ,congenital, hereditary, and neonatal diseases and abnormalities ,Fibrillin-1 ,Induced Pluripotent Stem Cells ,Germ layer ,Gene mutation ,Peripheral blood mononuclear cell ,Marfan Syndrome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,skin and connective tissue diseases ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Gene ,biology ,Cell Biology ,General Medicine ,biology.organism_classification ,medicine.disease ,Molecular biology ,Sendai virus ,030104 developmental biology ,lcsh:Biology (General) ,Mutation ,Leukocytes, Mononuclear ,Female ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Marfan syndrome (MFS) is a connective-tissue disorder caused mainly by heterozygous mutations in the FBN1 gene that encodes fibrillin-1. In this study, human induced pluripotent stem cell (iPSC) line ZZUSAHi003-A was generated from peripheral blood mononuclear cells (PBMCs) isolated from a female patient with MFS using non-integrative Sendai virus. The iPSC line carried the FBN1 gene mutation, showed the normal karyotype, expressed pluripotency markers and had the capacity to differentiate into three germ layers in vivo. This iPS line, ZZUSAHi003-A, could serve as a useful tool for studying pathogenic mechanisms of MFS.
- Published
- 2020
50. Establishment of an induced pluripotent stem cell line (WMUi016-A) from a patient with X-linked Dent disease (X-Dent) carrying the hemizygote mutation p.R718* (c.2152C T) in the CLCN5 gene
- Author
-
Jiajia Li, Maoping Chu, Dexuan Wang, Xing Rong, Xiaoling Guo, Congde Chen, Zhou Weizhong, Qian Weite, Huihui Chen, Zhe Zhang, Chao Li, Yinjuan Ding, and Hongfei Tong
- Subjects
Male ,Induced Pluripotent Stem Cells ,Dent Disease ,Gene mutation ,medicine.disease_cause ,Sendai virus ,medicine ,Humans ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Gene ,Hemizygote ,Mutation ,biology ,CLCN5 ,Cell Biology ,General Medicine ,Molecular biology ,lcsh:Biology (General) ,Child, Preschool ,biology.protein ,Reprogramming ,Developmental Biology - Abstract
The gene mutations of the chloride channel gene (CLCN5) can lead to the inherited X-linked Dent disease (X-Dent). The urine cells of a 4-year-old male X-Dent patient with the hemizygous CLCN5 gene mutation p.R718* (c.2152C > T) were reprogrammed into induced pluripotent stem cells (iPSCs) using integration free Sendai virus reprogramming system. The generated iPSCs stably expressed pluripotent stem cell markers and can be induced to differentiate into three germ layers in vitro. The karyotype of the generated iPSCs was normal (46, XY).
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.