Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Noncoding RNA ; 8(6)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36412910

ABSTRACT

Numerous noninvasive methods are currently being used to determine biomarkers for diseases such as cancer. However, these methods are not always precise and reliable. Thus, there is an unmet need for better diagnostic and prognostic biomarkers that will be used to diagnose cancer in early, more treatable stages of the disease. Exosomes are extracellular vesicles of endocytic origin released by the majority of cells. Exosomes contain and transport nucleic acids, proteins, growth factors, and cytokines from their parent cells to surrounding or even distant cells via circulation in biofluids. Exosomes have attracted the interest of researchers, as recent data indicate that exosome content may be indicative of disease stages and may contribute to disease progression via exosome-mediated extracellular communication. Therefore, the contents of these vesicles are being investigated as possible biomarkers for disease diagnosis and prognosis. The functions of exosomes and their contents in disease development are becoming clearer as isolation and analytical methods, such as RNA sequencing, advance. In this review, we discuss current advances and challenges in exosomal content analyses with emphasis on information that can be generated using RNA sequencing. We also discuss how the RNA sequencing of exosomes may be used to discover novel biomarkers for the detection of different stages for various cancers using specific microRNAs that were found to be differentially expressed between healthy controls and cancer-diagnosed subjects.

2.
PLoS One ; 17(4): e0266179, 2022.
Article in English | MEDLINE | ID: mdl-35446871

ABSTRACT

Mesenchymal stromal cells (MSCs) can be utilized clinically for treatment of conditions that result from excessive inflammation. In a pro-inflammatory environment, MSCs adopt an anti-inflammatory phenotype resulting in immunomodulation. A sub-type of MSCs referred to as "marrow-isolated adult multilineage inducible" (MIAMI) cells, which were isolated from bone marrow, were utilized to show that the addition of autophagy modulators, tamoxifen (TX) or chloroquine (CQ), can alter how MIAMI cells respond to IFNγ exposure in vitro resulting in an increased immunoregulatory capacity of the MIAMI cells. Molecularly, it was also shown that TX and CQ each alter both the levels of immunomodulatory genes and microRNAs which target such genes. However, the role of other non-coding RNAs (ncRNAs) such as long non-coding RNAs (lncRNAs) in regulating the response of MSCs to inflammation has been poorly studied. Here, we utilized transcriptomics and data mining to analyze the putative roles of various differentially regulated lncRNAs in MIAMI cells exposed to IFNγ with (or without) TX or CQ. The aim of this study was to investigate how the addition of TX and CQ alters lncRNA levels and evaluate how such changes could alter previously observed TX- and CQ-driven changes to the immunomodulatory properties of MIAMI cells. Data analysis revealed 693 long intergenic non-coding RNAS (lincRNAs), 480 pseudogenes, and 642 antisense RNAs that were differentially regulated with IFNγ, IFNγ+TX and IFNγ+CQ treatments. Further analysis of these RNA species based on the existing literature data revealed 6 antisense RNAs, 2 pseudogenes, and 5 lincRNAs that have the potential to modulate MIAMI cell's response to IFNγ treatment. Functional analysis of these genomic species based on current literature linking inflammatory response and ncRNAs indicated their potential for regulation of several key pro- and anti-inflammatory responses, including NFκB signaling, cytokine secretion and auto-immune responses. Overall, this work found potential involvement of multiple pro-and anti-inflammatory pathways and molecules in modulating MIAMI cells' response to inflammation.


Subject(s)
RNA, Long Noncoding , Autophagy , Chloroquine/pharmacology , Humans , Inflammation/genetics , Interferon-gamma/pharmacology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tamoxifen/pharmacology
3.
Noncoding RNA Res ; 7(1): 7-15, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35087990

ABSTRACT

The field of extracellular vesicles has been rapidly developing after it became evident that a defined subset of vesicles, called exosomes, can modulate several biological functions in distant cells and tissues. Exosomes range in a size from 40 to 160 nm in diameter, are released by majority of cells in our body, and carry molecules which reflect the cell of origin. The types of biomolecules packed, their respective purpose, and their impact on the physiological state of distinct cells and tissues should be understood to advance the using of exosomes as biomarkers of health and disease. Many of such physiological effects can be linked to exosomal RNA molecules which include both coding and non-coding RNAs. The biological role(s) of various exosomal RNAs have started being recognized after RNA sequencing methods became widely available which led to discovery of a variety of RNA molecules in exosomes and their roles in regulating of many biological processes are beginning to be unraveled. In present review, we outline and discuss recent progress in the elucidation of the various biological processes driven by exosomal RNA and their relevance for several major conditions including disorders of central nervous system, cardiovascular system, metabolism, cancer, and immune system. Furthermore, we also discuss potential use of exosomes as valuable therapeutics for tissue regeneration and for conditions resulting from excessive inflammation. While exosome research is still in its infancy, in-depth understanding of exosome formation, their biological effects, and specific cell-targeting will uncover how they can be used as disease biomarkers and therapeutics.

4.
PLoS One ; 16(1): e0246112, 2021.
Article in English | MEDLINE | ID: mdl-33481943

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0239238.].

5.
Stem Cell Res Ther ; 11(1): 489, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33208173

ABSTRACT

Over the last decades, the cancer survival rate has increased due to personalized therapies, the discovery of targeted therapeutics and novel biological agents, and the application of palliative treatments. Despite these advances, tumor resistance to chemotherapy and radiation and rapid progression to metastatic disease are still seen in many patients. Evidence has shown that cancer stem cells (CSCs), a sub-population of cells that share many common characteristics with somatic stem cells (SSCs), contribute to this therapeutic failure. The most critical properties of CSCs are their self-renewal ability and their capacity for differentiation into heterogeneous populations of cancer cells. Although CSCs only constitute a low percentage of the total tumor mass, these cells can regrow the tumor mass on their own. Initially identified in leukemia, CSCs have subsequently been found in cancers of the breast, the colon, the pancreas, and the brain. Common genetic and phenotypic features found in both SSCs and CSCs, including upregulated signaling pathways such as Notch, Wnt, Hedgehog, and TGF-ß. These pathways play fundamental roles in the development as well as in the control of cell survival and cell fate and are relevant to therapeutic targeting of CSCs. The differences in the expression of membrane proteins and exosome-delivered microRNAs between SSCs and CSCs are also important to specifically target the stem cells of the cancer. Further research efforts should be directed toward elucidation of the fundamental differences between SSCs and CSCs to improve existing therapies and generate new clinically relevant cancer treatments.


Subject(s)
Adult Stem Cells , Neoplasms , Cell Differentiation , Humans , Neoplasms/genetics , Neoplasms/therapy , Neoplastic Stem Cells , Signal Transduction
6.
PLoS One ; 15(9): e0239238, 2020.
Article in English | MEDLINE | ID: mdl-32941515

ABSTRACT

Zika virus (ZIKV) is a single-stranded RNA virus belonging to the family Flaviviridae. ZIKV predominantly enters cells using the TAM-family protein tyrosine kinase receptor AXL, which is expressed on a range of cell types, including neural progenitor cells, keratinocytes, dendritic cells, and osteoblasts. ZIKV infections have been associated with fetal brain damage, which prompted the World Health Organization to declare a public health emergency in 2016. ZIKV infection has also been linked to birth defects in other organs. Several studies have reported congenital heart defects (CHD) in ZIKV infected infants and cardiovascular complications in adults infected with ZIKV. To develop a better understanding of potential causes for these pathologies at a cellular level, we characterized ZIKV infection of human fetal cardiac mesenchymal stromal cells (fcMSCs), a cell type that is known to contribute to both embryological development as well as adult cardiac physiology. Total RNA, supernatants, and/or cells were collected at various time points post-infection to evaluate ZIKV replication, cell death, and antiviral responses. We found that ZIKV productively infected fcMSCs with peak (~70%) viral mRNA detected at 48 h. Use of an antibody blocking the AXL receptor decreased ZIKV infection (by ~50%), indicating that the receptor is responsible to a large extent for viral entry into the cell. ZIKV also altered protein expression of several mesenchymal cell markers, which suggests that ZIKV could affect fcMSCs' differentiation process. Gene expression analysis of fcMSCs exposed to ZIKV at 6, 12, and 24 h post-infection revealed up-regulation of genes/pathways associated with interferon-stimulated antiviral responses. Stimulation of TLR3 (using poly I:C) or TLR7 (using Imiquimod) prior to ZIKV infection suppressed viral replication in a dose-dependent manner. Overall, fcMSCs can be a target for ZIKV infection, potentially resulting in CHD during embryological development and/or cardiovascular issues in ZIKV infected adults.


Subject(s)
Human Embryonic Stem Cells/virology , Mesenchymal Stem Cells/virology , Myocytes, Cardiac/virology , Virus Replication , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Cell Death , Cells, Cultured , Chlorocebus aethiops , Human Embryonic Stem Cells/metabolism , Humans , Interferons/genetics , Interferons/metabolism , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Vero Cells , Zika Virus/pathogenicity , Zika Virus Infection/metabolism , Axl Receptor Tyrosine Kinase
7.
Stem Cell Res Ther ; 10(1): 395, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852519

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs), adult stromal cells most commonly isolated from bone marrow (BM), are being increasingly utilized in various therapeutic applications including tissue repair via immunomodulation, which is recognized as one of their most relevant mechanism of action. The promise of MSC-based therapies is somewhat hindered by their apparent modest clinical benefits, highlighting the need for approaches that would increase the efficacy of such therapies. Manipulation of cellular stress-response mechanism(s) such as autophagy, a catabolic stress-response mechanism, with small molecules prior to or during MSC injection could improve MSCs' therapeutic efficacy. Unfortunately, limited information exists on how manipulation of autophagy affects MSCs' response to inflammation and subsequent immunoregulatory properties. METHODS: In this study, we exposed BM-MSC precursor cells, "marrow-isolated adult multilineage inducible" (MIAMI) cells, to autophagy modulators tamoxifen (TX) or chloroquine (CQ), together with IFN-γ. Exposed cells then underwent RNA sequencing (RNAseq) to determine the effects of TX or CQ co-treatments on cellular response to IFN-γ at a molecular level. Furthermore, we evaluated their immunoregulatory capacity using activated CD4+ T cells by analyzing T cell activation marker CD25 and the percentage of proliferating T cells after co-culturing the cells with MIAMI cells treated or not with TX or CQ. RESULTS: RNAseq data indicate that the co-treatments alter both mRNA and protein levels of key genes responsible for MSCs' immune-regulatory properties. Interestingly, TX and CQ also altered some of the microRNAs targeting such key genes. In addition, while IFN-γ treatment alone increased the surface expression of PD-L1 and secretion of IDO, this increase was further enhanced with TX. An improvement in MIAMI cells' ability to decrease the activation and proliferation of T cells was also observed with TX, and to a lesser extent, CQ co-treatments. CONCLUSION: Altogether, this work suggests that both TX and CQ have a potential to enhance MIAMI cells' immunoregulatory properties. However, this enhancement is more pronounced with TX co-treatment.


Subject(s)
Cell Proliferation/drug effects , Chloroquine/pharmacology , Interferon-gamma/pharmacology , Tamoxifen/pharmacology , Autophagy/drug effects , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Coculture Techniques , Gene Expression/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-6/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism
8.
Stem Cell Res Ther ; 10(1): 371, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31801632

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs), due to their regenerative and immunomodulatory properties, are therapeutically used for diseases, including heart failure. As early gestational-phase embryonic tissues exhibit extraordinary regenerative potential, fetal MSCs exposed to inflammation offer a unique opportunity to evaluate molecular mechanisms underlying preferential healing, and investigate their inherent abilities to communicate with the immune system during development. The principal aim of this study was to evaluate the effects of interferon-γ (IFNγ) on the immunomodulatory effects of first-trimester human fetal cardiac (hfc)-MSCs. METHODS: hfcMSCs (gestational week 8) were exposed to IFNγ, with subsequent analysis of the whole transcriptome, based on RNA sequencing. Exploration of surface-expressed immunoregulatory mediators and modulation of T cell responses were performed by flow cytometry. Presence and activity of soluble mediators were assessed by ELISA or high-performance liquid chromatography. RESULTS: Stimulation of hfcMSCs with IFNγ revealed significant transcriptional changes, particularly in respect to the expression of genes belonging to antigen presentation pathways, cell cycle control, and interferon signaling. Expression of immunomodulatory genes and associated functional changes, including indoleamine 2,3-dioxygenase activity, and regulation of T cell activation and proliferation via programmed cell death protein (PD)-1 and its ligands PD-L1 and PD-L2, were significantly upregulated. These immunoregulatory molecules diminished rapidly upon withdrawal of inflammatory stimulus, indicating a high degree of plasticity by hfcMSCs. CONCLUSIONS: To our knowledge, this is the first study performing a systematic evaluation of inflammatory responses and immunoregulatory properties of first-trimester cardiac tissue. In summary, our study demonstrates the dynamic responsiveness of hfcMSCs to inflammatory stimuli. Further understanding as to the immunoregulatory properties of hfcMSCs may be of benefit in the development of novel stromal cell therapeutics for cardiovascular disease.


Subject(s)
Immunomodulation/drug effects , Interferon-gamma/pharmacology , Transcriptome/drug effects , Apoptosis/drug effects , B7-H1 Antigen/metabolism , Cell Proliferation , Fetus/cytology , HLA Antigens/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphocyte Activation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Receptors, Interferon/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Interferon gamma Receptor
9.
Cytotherapy ; 21(4): 380-392, 2019 04.
Article in English | MEDLINE | ID: mdl-30876741

ABSTRACT

Stem cells (SCs) have been proven to possess regenerative and immunomodulatory properties and can be used to treat diseases that involve loss of cells due to tissue damage or inflammation. For this approach to succeed, SCs or their derivatives should be able to engraft in the target tissue at least for a short period of time. Unfortunately, once injected, therapeutic SCs will encounter a hostile environment, including hypoxia, lack of nutrients and stromal support, and cells may also be targeted and rejected by the immune system. Therefore, SC's stress-response mechanisms likely play a significant role in survival of injected cells and possibly contribute to their therapeutic efficacy. Autphagy, a stress-response pathway, is involved in many different cellular processes, such as survival during hypoxia and nutrient deprivation, cellular differentiation and de-differentiation, and it can also contribute to their immunovisibility by regulating antigen presentation and cytokine secretion. Autophagy machinery interacts with many proteins and signaling pathways that regulate SC properties, including PI3K/Akt, mammalian target of rapamycin (mTOR), Wnt, Hedgehog and Notch, and it is also involved in regulating intracellular reactive oxygen species (ROS) levels. In this review, we contend that autophagy is an important therapeutic target that can be used to improve the outcome of SC-based tissue repair and regeneration. Further research should reveal whether inhibition or stimulation of autophagy increases the therapeutic utility of SCs and it should also identify appropriate therapeutic regimens that can be applied in the clinic.


Subject(s)
Autophagy , Stem Cell Transplantation , Stem Cells/cytology , Animals , Cell Differentiation , Extracellular Matrix/metabolism , Humans , Signal Transduction
10.
DNA Cell Biol ; 38(6): 561-571, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30920300

ABSTRACT

Gulf War Illness (GWI) affects about 25% of Persian Gulf veterans with a cluster of chronic symptoms, including immune dysfunction and neurological issues. Recent studies implicate gene expression changes in immune function to be associated with GWI. Since DNA methylation can regulate such changes in gene expression, and disruption of DNA methylation pattern is implicated in various immune and neurological diseases, we aimed to study the DNA methylation patterns in peripheral blood mononuclear cells from GWI patients. Global DNA methylation levels were similar in GWI patients and controls. However, the genome-wide microarray technology detected 10,767 differentially methylated CpG sites across gene regulatory elements and within coding regions. Approximately 88% of them were hypermethylated in GWI patients. The separate analysis found 776 differentially methylated gene promoters (DMP), which were predominantly hypermethylated. Pyrosequencing validation confirmed microarray results. Functional analysis revealed that majority of the DMPs belonged to genes responsible for metabolism and immune system. This is the first pilot human study characterizing genome-wide epigenetic changes associated with GWI. It suggests a significant contribution of epigenetic dysfunction in GWI. Moreover, it supports the dysregulation of immune function in GWI. Lastly, it suggests studies with the larger cohort to validate our findings.


Subject(s)
DNA Methylation , Persian Gulf Syndrome/genetics , Adult , Cohort Studies , CpG Islands , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Persian Gulf Syndrome/immunology , Pilot Projects , Promoter Regions, Genetic , Sequence Analysis, DNA
11.
Cancers (Basel) ; 11(1)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30634595

ABSTRACT

Natural killer (NK) cells stand out as promising candidates for cellular immunotherapy due to their capacity to kill malignant cells. However, the therapeutic use of NK cells is often dependent on cell expansion and activation with considerable amounts of serum and exogenous cytokines. We aimed to develop an expansion protocol for NK-92 cells in an effort to generate a cost-efficient, xeno-free, clinical grade manufactured master cell line for therapeutic applications. By making functional assays with NK-92 cells cultured under serum-free conditions (NK-92SF) and comparing to serum-supplemented NK-92 cells (NK-92S) we did not observe significant alterations in the viability, proliferation, receptor expression levels, or in perforin and granzyme levels. Interestingly, even though NK-92SF cells displayed decreased degranulation and cytotoxicity against tumor cells in vitro, the degranulation capacity was recovered after overnight incubation with 20% serum in the medium. Moreover, lentiviral vector-based genetic modification efficiency of NK-92SF cells was comparable with NK-92S cells. The application of similar strategies can be useful in reducing the costs of manufacturing cells for clinical use and can help us understand and implement strategies towards chemically defined expansion and genetic modification protocols.

12.
PLoS One ; 13(7): e0201066, 2018.
Article in English | MEDLINE | ID: mdl-30036399

ABSTRACT

BACKGROUND: Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a complex condition involving multiple organ systems and characterized by persistent/relapsing debilitating fatigue, immune dysfunction, neurological problems, and other symptoms not curable for at least 6 months. Disruption of DNA methylation patterns has been tied to various immune and neurological diseases; however, its status in ME/CFS remains uncertain. Our study aimed at identifying changes in the DNA methylation patterns that associate with ME/CFS. METHODS: We extracted genomic DNA from peripheral blood mononuclear cells from 13 ME/CFS study subjects and 12 healthy controls and measured global DNA methylation by ELISA-like method and site-specific methylation status using Illumina MethylationEPIC microarrays. Pyrosequencing validation included 33 ME/CFS cases and 31 controls from two geographically distant cohorts. RESULTS: Global DNA methylation levels of ME/CFS cases were similar to those of controls. However, microarray-based approach allowed detection of 17,296 differentially methylated CpG sites in 6,368 genes across regulatory elements and within coding regions of genes. Analysis of DNA methylation in promoter regions revealed 307 differentially methylated promoters. Ingenuity pathway analysis indicated that genes associated with differentially methylated promoters participated in at least 15 different pathways mostly related to cell signaling with a strong immune component. CONCLUSIONS: This is the first study that has explored genome-wide epigenetic changes associated with ME/CFS using the advanced Illumina MethylationEPIC microarrays covering about 850,000 CpG sites in two geographically distant cohorts of ME/CFS cases and matched controls. Our results are aligned with previous studies that indicate a dysregulation of the immune system in ME/CFS. They also suggest a potential role of epigenetic de-regulation in the pathobiology of ME/CFS. We propose screening of larger cohorts of ME/CFS cases to determine the external validity of these epigenetic changes in order to implement them as possible diagnostic markers in clinical setting.


Subject(s)
DNA Methylation , Fatigue Syndrome, Chronic/metabolism , Cohort Studies , CpG Islands , Epigenesis, Genetic , Fatigue Syndrome, Chronic/genetics , Female , Humans , Microarray Analysis , Middle Aged , Promoter Regions, Genetic
13.
Mol Ther ; 25(8): 1900-1916, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28527723

ABSTRACT

Oncolytic viruses (OVs) offer a promising therapeutic approach to treat multiple types of cancer. In this study, we show that the manipulation of the antioxidant network via transcription factor Nrf2 augments vesicular stomatitis virus Δ51 (VSVΔ51) replication and sensitizes cancer cells to viral oncolysis. Activation of Nrf2 signaling by the antioxidant compound sulforaphane (SFN) leads to enhanced VSVΔ51 spread in OV-resistant cancer cells and improves the therapeutic outcome in different murine syngeneic and xenograft tumor models. Chemoresistant A549 lung cancer cells that display constitutive dominant hyperactivation of Nrf2 signaling are particularly vulnerable to VSVΔ51 oncolysis. Mechanistically, enhanced Nrf2 signaling stimulated viral replication in cancer cells and disrupted the type I IFN response via increased autophagy. This study reveals a previously unappreciated role for Nrf2 in the regulation of autophagy and the innate antiviral response that complements the therapeutic potential of VSV-directed oncolysis against multiple types of OV-resistant or chemoresistant cancer.


Subject(s)
Autophagy , NF-E2-Related Factor 2/metabolism , Oncolytic Viruses/physiology , Signal Transduction , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/physiology , Animals , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Autophagy/drug effects , Cell Line , Combined Modality Therapy , Disease Models, Animal , Host-Pathogen Interactions/immunology , Humans , Immunity/drug effects , Immunity, Innate/drug effects , Isothiocyanates/pharmacology , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Virotherapy , Sequence Deletion , Signal Transduction/drug effects , Sulfoxides , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/drug effects , Viral Matrix Proteins/genetics , Virus Replication/drug effects
14.
J Virol ; 89(20): 10612-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269188

ABSTRACT

UNLABELLED: The molecular interaction between viral RNA and the cytosolic sensor RIG-I represents the initial trigger in the development of an effective immune response against infection with RNA viruses, resulting in innate immune activation and subsequent induction of adaptive responses. In the present study, the adjuvant properties of a sequence-optimized 5'-triphosphate-containing RNA (5'pppRNA) RIG-I agonist (termed M8) were examined in combination with influenza virus-like particles (VLP) (M8-VLP) expressing H5N1 influenza virus hemagglutinin (HA) and neuraminidase (NA) as immunogens. In combination with VLP, M8 increased the antibody response to VLP immunization, provided VLP antigen sparing, and protected mice from a lethal challenge with H5N1 influenza virus. M8-VLP immunization also led to long-term protective responses against influenza virus infection in mice. M8 adjuvantation of VLP increased endpoint and antibody titers and inhibited influenza virus replication in lungs compared with approved or experimental adjuvants alum, AddaVax, and poly(I·C). Uniquely, immunization with M8-VLP stimulated a TH1-biased CD4 T cell response, as determined by increased TH1 cytokine levels in CD4 T cells and increased IgG2 levels in sera. Collectively, these data demonstrate that a sequence-optimized, RIG-I-specific agonist is a potent adjuvant that can be utilized to increase the efficacy of influenza VLP vaccination and dramatically improve humoral and cellular mediated protective responses against influenza virus challenge. IMPORTANCE: The development of novel adjuvants to increase vaccine immunogenicity is an important goal that seeks to improve vaccine efficacy and ultimately prevent infections that endanger human health. This proof-of-principle study investigated the adjuvant properties of a sequence-optimized 5'pppRNA agonist (M8) with enhanced capacity to stimulate antiviral and inflammatory gene networks using influenza virus-like particles (VLP) expressing HA and NA as immunogens. Vaccination with VLP in combination with M8 increased anti-influenza virus antibody titers and protected animals from lethal influenza virus challenge, highlighting the potential clinical use of M8 as an adjuvant in vaccine development. Altogether, the results describe a novel immunostimulatory agonist targeted to the cytosolic RIG-I sensor as an attractive vaccine adjuvant candidate that can be used to increase vaccine efficacy, a pressing issue in children and the elderly population.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/biosynthesis , DEAD-box RNA Helicases/immunology , Influenza Vaccines/immunology , Oligoribonucleotides/administration & dosage , Orthomyxoviridae Infections/prevention & control , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/genetics , Animals , DEAD Box Protein 58 , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Female , HEK293 Cells , Hemagglutinins, Viral/chemistry , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunization , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Mice, Inbred BALB C , Neuraminidase/chemistry , Neuraminidase/genetics , Neuraminidase/immunology , Oligoribonucleotides/genetics , Oligoribonucleotides/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Primary Cell Culture , Receptors, Immunologic , Survival Analysis , Th1-Th2 Balance/drug effects , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics
15.
Biol Chem ; 396(12): 1269-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26068902

ABSTRACT

Resistance to both cytotoxic and targeted therapies is a major problem facing cancer treatment. The mechanisms of resistance to unrelated drugs share many common features, including up-regulation of detoxifying pathways, activation of pro-survival mechanisms, and ineffective induction of cell death. Oncolytic viruses (OVs) are promising biotherapeutics for cancer treatment that specifically replicate in and lyse cancer cells. In addition to direct viral lysis, the anti-tumor effects of OVs are mediated via innate and adaptive immune responses, and several adaptation mechanisms such as autophagy appear to contribute to their anti-tumor properties. Autophagy is a versatile pathway that plays a key role in cancer survival during stressful conditions such as starvation or cytotoxic drug challenges. Autophagy also plays a role in mediating innate and adaptive immune responses by contributing to antigen presentation and cytokine secretion. This role of autophagy in regulation of immune responses can be utilized to design therapeutic combinations using approaches that either stimulate or block autophagy to potentiate therapeutic efficacy of OVs. Additional studies are needed to determine optimal multimodal combination approaches that will facilitate future successful clinical implementation of OV-based therapies.


Subject(s)
Autophagy/physiology , Drug Resistance , Neoplasms/therapy , Neoplasms/virology , Oncolytic Viruses , Humans , Neoplasms/drug therapy , T-Lymphocytes/immunology
16.
J Virol ; 89(15): 8011-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26018150

ABSTRACT

UNLABELLED: The cytosolic RIG-I (retinoic acid-inducible gene I) receptor plays a pivotal role in the initiation of the immune response against RNA virus infection by recognizing short 5'-triphosphate (5'ppp)-containing viral RNA and activating the host antiviral innate response. In the present study, we generated novel 5'ppp RIG-I agonists of varieous lengths, structures, and sequences and evaluated the generation of the antiviral and inflammatory responses in human epithelial A549 cells, human innate immune primary cells, and murine models of influenza and chikungunya viral pathogenesis. A 99-nucleotide, uridine-rich hairpin 5'pppRNA termed M8 stimulated an extensive and robust interferon response compared to other modified 5'pppRNA structures, RIG-I aptamers, or poly(I·C). Interestingly, manipulation of the primary RNA sequence alone was sufficient to modulate antiviral activity and inflammatory response, in a manner dependent exclusively on RIG-I and independent of MDA5 and TLR3. Both prophylactic and therapeutic administration of M8 effectively inhibited influenza virus and dengue virus replication in vitro. Furthermore, multiple strains of influenza virus that were resistant to oseltamivir, an FDA-approved therapeutic treatment for influenza, were highly sensitive to inhibition by M8. Finally, prophylactic M8 treatment in vivo prolonged survival and reduced lung viral titers of mice challenged with influenza virus, as well as reducing chikungunya virus-associated foot swelling and viral load. Altogether, these results demonstrate that 5'pppRNA can be rationally designed to achieve a maximal RIG-I-mediated protective antiviral response against human-pathogenic RNA viruses. IMPORTANCE: The development of novel therapeutics to treat human-pathogenic RNA viral infections is an important goal to reduce spread of infection and to improve human health and safety. This study investigated the design of an RNA agonist with enhanced antiviral and inflammatory properties against influenza, dengue, and chikungunya viruses. A novel, sequence-dependent, uridine-rich RIG-I agonist generated a protective antiviral response in vitro and in vivo and was effective at concentrations 100-fold lower than prototype sequences or other RNA agonists, highlighting the robust activity and potential clinical use of the 5'pppRNA against RNA virus infection. Altogether, the results identify a novel, sequence-specific RIG-I agonist as an attractive therapeutic candidate for the treatment of a broad range of RNA viruses, a pressing issue in which a need for new and more effective options persists.


Subject(s)
Chikungunya virus/immunology , DEAD-box RNA Helicases/immunology , Dengue Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , RNA, Viral/agonists , RNA, Viral/immunology , Virus Diseases/immunology , Animals , Cell Line , Chikungunya virus/chemistry , Chikungunya virus/genetics , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dengue Virus/chemistry , Dengue Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/genetics , Mice , Mice, Inbred BALB C , Nucleic Acid Conformation , RNA, Viral/genetics , Receptors, Immunologic , Virus Diseases/genetics , Virus Diseases/virology
17.
PLoS Pathog ; 10(12): e1004566, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25521078

ABSTRACT

Dengue virus (DENV) is a re-emerging arthropod borne flavivirus that infects more than 300 million people worldwide, leading to 50,000 deaths annually. Because dendritic cells (DC) in the skin and blood are the first target cells for DENV, we sought to investigate the early molecular events involved in the host response to the virus in primary human monocyte-derived dendritic cells (Mo-DC). Using a genome-wide transcriptome analysis of DENV2-infected human Mo-DC, three major responses were identified within hours of infection - the activation of IRF3/7/STAT1 and NF-κB-driven antiviral and inflammatory networks, as well as the stimulation of an oxidative stress response that included the stimulation of an Nrf2-dependent antioxidant gene transcriptional program. DENV2 infection resulted in the intracellular accumulation of reactive oxygen species (ROS) that was dependent on NADPH-oxidase (NOX). A decrease in ROS levels through chemical or genetic inhibition of the NOX-complex dampened the innate immune responses to DENV infection and facilitated DENV replication; ROS were also essential in driving mitochondrial apoptosis in infected Mo-DC. In addition to stimulating innate immune responses to DENV, increased ROS led to the activation of bystander Mo-DC which up-regulated maturation/activation markers and were less susceptible to viral replication. We have identified a critical role for the transcription factor Nrf2 in limiting both antiviral and cell death responses to the virus by feedback modulation of oxidative stress. Silencing of Nrf2 by RNA interference increased DENV-associated immune and apoptotic responses. Taken together, these data demonstrate that the level of oxidative stress is critical to the control of both antiviral and apoptotic programs in DENV-infected human Mo-DC and highlight the importance of redox homeostasis in the outcome of DENV infection.


Subject(s)
Apoptosis/physiology , Dendritic Cells/physiology , Dendritic Cells/virology , Dengue Virus/physiology , Immunity, Innate/physiology , Oxidative Stress/physiology , Cells, Cultured , Dendritic Cells/pathology , Gene Expression Profiling , Humans , In Vitro Techniques , Interferon Regulatory Factor-3/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/metabolism , Virus Replication/physiology
18.
J Virol ; 88(5): 2927-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371063

ABSTRACT

Vesicular stomatitis virus (VSV) is an oncolytic virus that induces cancer cell death through activation of the apoptotic pathway. Intrinsic resistance to oncolysis is found in some cell lines and many primary tumors as a consequence of residual innate immunity to VSV. In resistant-tumor models, VSV oncolytic potential can be reversibly stimulated by combination with epigenetic modulators, such as the histone deacetylase inhibitor vorinostat. Based on this reversible effect of vorinostat, we reasoned that critical host genes involved in oncolysis may likewise be reversibly regulated by vorinostat. A transcriptome analysis in prostate cancer PC3 cells identified a subset of NF-κB target genes reversibly regulated by vorinostat, as well as a group of interferon (IFN)-stimulated genes (ISGs). Consistent with the induction of NF-κB target genes, vorinostat-mediated enhancement of VSV oncolysis increased hyperacetylation of NF-κB RELA/p65. Additional bioinformatics analysis revealed that NF-κB signaling also increased the expression of several autophagy-related genes. Kinetically, autophagy preceded apoptosis, and apoptosis was observed only when cells were treated with both VSV and vorinostat. VSV replication and cell killing were suppressed when NF-κB signaling was inhibited using pharmacological or genetic approaches. Inhibition of autophagy by 3-methyladenine (3-MA) enhanced expression of ISGs, and either 3-MA treatment or genetic ablation of the autophagic marker Atg5 decreased VSV replication and oncolysis. Together, these data demonstrate that vorinostat stimulates NF-κB activity in a reversible manner via modulation of RELA/p65 signaling, leading to induction of autophagy, suppression of the IFN-mediated response, and subsequent enhancement of VSV replication and apoptosis.


Subject(s)
Autophagy , Histone Deacetylase Inhibitors/pharmacology , NF-kappa B/metabolism , Oncolytic Viruses/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Vesicular stomatitis Indiana virus/drug effects , Acetylation , Animals , Autophagy/drug effects , Cell Line, Tumor , Chromatin/metabolism , Cluster Analysis , Gene Knockdown Techniques , Humans , Hydroxamic Acids/pharmacology , Male , Mice , NF-kappa B/antagonists & inhibitors , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Prostatic Neoplasms/therapy , Protein Binding , Protein Transport/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcriptome , Vesicular stomatitis Indiana virus/genetics , Virus Replication , Vorinostat
19.
Mol Ther ; 21(7): 1413-23, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23689597

ABSTRACT

Many primary cancers including chronic lymphocytic leukemia (CLL) are resistant to vesicular stomatitis virus (VSV)-induced oncolysis due to overexpression of the antiapoptotic and antiautophagic members of the B-cell lymphoma-2 (BCL-2) family. In the present study, we investigated the mechanisms of CLL cell death induced as a consequence of VSV infection in the presence of BCL-2 inhibitors, obatoclax, and ABT-737 in primary ex vivo CLL patient samples. Microarray analysis of primary CD19⁺ CD5⁺ CLL cells treated with obatoclax and VSV revealed changes in expression of genes regulating apoptosis, the mechanistic target of rapamycin (mTOR) pathway, and cellular metabolism. A combined therapeutic effect was observed for VSV and BCL-2 inhibitors in cells from untreated patients and from patients unresponsive to standard of care therapy. In addition, combination treatment induced several markers of autophagy--LC3-II accumulation, p62 degradation, and staining of autophagic vacuoles. Inhibition of early stage autophagy using 3-methyladenine (3-MA) led to increased apoptosis in CLL samples. Mechanistically, a combination of BCL-2 inhibitors and VSV disrupted inhibitory interactions of Beclin-1 with BCL-2 and myeloid cell leukemia-1 (MCL-1), thus biasing cells toward autophagy. We propose a mechanism in which changes in cellular metabolism, coupled with pharmacologic disruption of the BCL-2-Beclin-1 interactions, facilitate induction of apoptosis and autophagy to mediate the cytolytic effect of VSV.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Oncolytic Viruses/physiology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Vesicular stomatitis Indiana virus/genetics , Animals , Biphenyl Compounds/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Indoles , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Nitrophenols/pharmacology , Oncolytic Viruses/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/pharmacology , Sulfonamides/pharmacology , Vesicular stomatitis Indiana virus/physiology
20.
Curr Opin Virol ; 2(5): 629-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22910124

ABSTRACT

Intrinsic and acquired drug resistance remains a fundamental obstacle to successful applications of anticancer therapies for lung cancer. Combining conventional therapies with immunotherapeutic approaches is a promising strategy to circumvent lung cancer drug resistance. Genetically modified oncolytic viruses (OVs) kill tumor cells via completely unique mechanisms compared to small molecule chemotherapeutics typically used in lung cancer treatment and can also be used to deliver specific toxic, therapeutic or immunomodulatory genes to tumor cells. Recent pre-clinical and clinical studies with oncolytic vaccine approaches have revealed promising combination strategies that enhance oncolysis of tumor cells and circumvent tumor resistance mechanisms. As clinical trials with oncolytic vaccines progress, and as the knowledge acquired from these studies builds a foundation demonstrating OVs safety and efficacy, novel combination approaches could soon have a major impact on the clinical management of patients diagnosed with lung cancer.


Subject(s)
Genetic Therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Oncolytic Viruses/physiology , Animals , Drug Resistance, Neoplasm , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Lung Neoplasms/metabolism , Oncolytic Viruses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...