Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937436

ABSTRACT

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Subject(s)
Antigens, Neoplasm , B7-H1 Antigen , CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Liver Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Tumor Microenvironment , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Animals , Oncolytic Virotherapy/methods , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/immunology , Tumor Microenvironment/immunology , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Humans , Liver Neoplasms/therapy , Liver Neoplasms/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Interferon-beta/metabolism , Interferon-beta/immunology , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , T-Lymphocytes/immunology , Female , Vesiculovirus/immunology , Vesiculovirus/genetics
2.
Res Sq ; 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38045348

ABSTRACT

Although immune checkpoint inhibition (ICI) has produced profound survival benefits in a broad variety of tumors, a proportion of patients do not respond. Treatment failure is in part due to immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, we developed a vesicular stomatitis virus expressing interferon-ß (VSV-IFNß) as a viro-immunotherapy against HCC. Since HCC standard of care atezolizumab/bevacizumab incorporates ICI, we tested the hypothesis that pro-inflammatory VSV-IFNß would recruit, prime, and activate anti-tumor T cells, whose activity anti-PD-L1 ICI would potentiate. However, in a partially anti-PD-L1-responsive model of HCC, addition of VSV-IFNß abolished anti-PD-L1 therapy. Cytometry by Time of Flight showed that VSV-IFNß expanded dominant anti-viral effector CD8 T cells with concomitant, relative disappearance of anti-tumor T cell populations which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, the potent anti-viral response became amalgamated with an anti-tumor T cell response generating highly significant cures compared to anti-PD-L1 ICI alone. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, by chimerizing anti-viral and anti-tumor T cell responses through encoding tumor antigens within the virus, oncolytic virotherapy can be purposed for very effective immune driven tumor clearance and can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.

3.
Front Immunol ; 14: 1279387, 2023.
Article in English | MEDLINE | ID: mdl-38022659

ABSTRACT

Introduction: Metastatic uveal melanoma (MUM) has a poor prognosis and treatment options are limited. These patients do not typically experience durable responses to immune checkpoint inhibitors (ICIs). Oncolytic viruses (OV) represent a novel approach to immunotherapy for patients with MUM. Methods: We developed an OV with a Vesicular Stomatitis Virus (VSV) vector modified to express interferon-beta (IFN-ß) and Tyrosinase Related Protein 1 (TYRP1) (VSV-IFNß-TYRP1), and conducted a Phase 1 clinical trial with a 3 + 3 design in patients with MUM. VSV-IFNß-TYRP1 was injected into a liver metastasis, then administered on the same day as a single intravenous (IV) infusion. The primary objective was safety. Efficacy was a secondary objective. Results: 12 patients with previously treated MUM were enrolled. Median follow up was 19.1 months. 4 dose levels (DLs) were evaluated. One patient at DL4 experienced dose limiting toxicities (DLTs), including decreased platelet count (grade 3), increased aspartate aminotransferase (AST), and cytokine release syndrome (CRS). 4 patients had stable disease (SD) and 8 patients had progressive disease (PD). Interferon gamma (IFNγ) ELIspot data showed that more patients developed a T cell response to virus encoded TYRP1 at higher DLs, and a subset of patients also had a response to other melanoma antigens, including gp100, suggesting epitope spreading. 3 of the patients who responded to additional melanoma antigens were next treated with ICIs, and 2 of these patients experienced durable responses. Discussion: Our study found that VSV-IFNß -TYRP1 can be safely administered via intratumoral (IT) and IV routes in a previously treated population of patients with MUM. Although there were no clear objective radiographic responses to VSV-IFNß-TYRP1, dose-dependent immunogenicity to TYRP1 and other melanoma antigens was seen.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Vesicular Stomatitis , Animals , Humans , Interferon-beta/metabolism , Melanoma-Specific Antigens , Monophenol Monooxygenase/metabolism , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , T-Lymphocytes/metabolism , Vesicular stomatitis Indiana virus
4.
Mol Ther Oncolytics ; 29: 129-142, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37313455

ABSTRACT

In multiple models of oncolytic virotherapy, it is common to see an early anti-tumor response followed by recurrence. We have previously shown that frontline treatment with oncolytic VSV-IFN-ß induces APOBEC proteins, promoting the selection of specific mutations that allow tumor escape. Of these mutations in B16 melanoma escape (ESC) cells, a C-T point mutation in the cold shock domain-containing E1 (CSDE1) gene was present at the highest frequency, which could be used to ambush ESC cells by vaccination with the mutant CSDE1 expressed within the virus. Here, we show that the evolution of viral ESC tumor cells harboring the escape-promoting CSDE1C-T mutation can also be exploited by a virological ambush. By sequential delivery of two oncolytic VSVs in vivo, tumors which would otherwise escape VSV-IFN-ß oncolytic virotherapy could be cured. This also facilitated the priming of anti-tumor T cell responses, which could be further exploited using immune checkpoint blockade with the CD200 activation receptor ligand (CD200AR-L) peptide. Our findings here are significant in that they offer the possibility to develop oncolytic viruses as highly specific, escape-targeting viro-immunotherapeutic agents to be used in conjunction with recurrence of tumors following multiple different types of frontline cancer therapies.

5.
Sci Transl Med ; 14(640): eabn2231, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35417192

ABSTRACT

Oncolytic viruses (OVs) encoding a variety of transgenes have been evaluated as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-modified T cells in the solid tumor microenvironment (TME). Here, using systemically delivered OVs and CAR T cells in immunocompetent mouse models, we have defined a mechanism by which OVs can potentiate CAR T cell efficacy against solid tumor models of melanoma and glioma. We show that stimulation of the native T cell receptor (TCR) with viral or virally encoded epitopes gives rise to enhanced proliferation, CAR-directed antitumor function, and distinct memory phenotypes. In vivo expansion of dual-specific (DS) CAR T cells was leveraged by in vitro preloading with oncolytic vesicular stomatitis virus (VSV) or reovirus, allowing for a further in vivo expansion and reactivation of T cells by homologous boosting. This treatment led to prolonged survival of mice with subcutaneous melanoma and intracranial glioma tumors. Human CD19 CAR T cells could also be expanded in vitro with TCR reactivity against viral or virally encoded antigens and was associated with greater CAR-directed cytokine production. Our data highlight the utility of combining OV and CAR T cell therapy and show that stimulation of the native TCR can be exploited to enhance CAR T cell activity and efficacy in mice.


Subject(s)
Glioma , Melanoma , Oncolytic Virotherapy , Oncolytic Viruses , Receptors, Chimeric Antigen , Animals , Glioma/therapy , Immunotherapy, Adoptive , Melanoma/therapy , Mice , Oncolytic Viruses/physiology , Receptors, Antigen, T-Cell , T-Lymphocytes , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Nat Commun ; 12(1): 1930, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33772027

ABSTRACT

In our clinical trials of oncolytic vesicular stomatitis virus expressing interferon beta (VSV-IFNß), several patients achieved initial responses followed by aggressive relapse. We show here that VSV-IFNß-escape tumors predictably express a point-mutated CSDE1P5S form of the RNA-binding Cold Shock Domain-containing E1 protein, which promotes escape as an inhibitor of VSV replication by disrupting viral transcription. Given time, VSV-IFNß evolves a compensatory mutation in the P/M Inter-Genic Region which rescues replication in CSDE1P5S cells. These data show that CSDE1 is a major cellular co-factor for VSV replication. However, CSDE1P5S also generates a neo-epitope recognized by non-tolerized T cells. We exploit this predictable neo-antigenesis to drive, and trap, tumors into an escape phenotype, which can be ambushed by vaccination against CSDE1P5S, preventing tumor escape. Combining frontline therapy with escape-targeting immunotherapy will be applicable across multiple therapies which drive tumor mutation/evolution and simultaneously generate novel, targetable immunopeptidomes associated with acquired treatment resistance.


Subject(s)
DNA-Binding Proteins/immunology , Interferon-beta/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , RNA-Binding Proteins/immunology , Vesicular stomatitis Indiana virus/immunology , Virus Replication/immunology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Immunotherapy/methods , Interferon-beta/metabolism , Mice, Inbred C57BL , Mutation , Oncolytic Viruses/metabolism , Oncolytic Viruses/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vesicular stomatitis Indiana virus/metabolism , Vesicular stomatitis Indiana virus/physiology
7.
Mol Ther ; 28(12): 2540-2552, 2020 12 02.
Article in English | MEDLINE | ID: mdl-32877695

ABSTRACT

Enhancing the immunogenicity of tumor-associated antigens would represent a major advance for anti-tumor vaccination strategies. Here, we investigated structure-directed antigen destabilization as a strategy to improve the degradation, immunogenic epitope presentation, and T cell activation against a vesicular stomatitis virus (VSV)-encoded tumor antigen. We used the crystal structure of the model antigen ovalbumin to identify charge-disrupting amino acid mutations that were predicted to decrease the stability of the protein. One mutation, OVA-C12R, significantly reduced the half-life of the protein and was preferentially degraded in a 26-S proteasomal-dependent manner. The destabilized ovalbumin protein exhibited enhanced presentation of the major histocompatibility complex (MHC) class I immunogenic epitope, SIINFEKL, on the surface of B16F10 cells or murine bone marrow-derived dendritic cells (BMDCs) in vitro. Enhanced presentation correlated with better recognition by cognate CD8 OT-I T cells as measured by activation, proliferation, and effector cytokine production. Finally, VSV encoding the degradation-prone antigen was better able to prime an antigen ovalbumin-specific CD8 T cell response in vivo without altering the anti-viral CD8 T cell response. Our studies highlight that not only is the choice of antigen in cancer vaccines of importance, but that emphasis should be placed on modifying antigen quality to ensure optimal priming of anti-tumor responses.


Subject(s)
Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/immunology , Immunity , Lymphocyte Activation , Ovalbumin/genetics , Vesiculovirus/genetics , Animals , Antigen Presentation , Antigens, Neoplasm/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Epitopes/immunology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncolytic Virotherapy/methods , Ovalbumin/chemistry , Protein Stability
8.
Nat Commun ; 11(1): 3187, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32581235

ABSTRACT

The application of adoptive T cell therapies, including those using chimeric antigen receptor (CAR)-modified T cells, to solid tumors requires combinatorial strategies to overcome immune suppression associated with the tumor microenvironment. Here we test whether the inflammatory nature of oncolytic viruses and their ability to remodel the tumor microenvironment may help to recruit and potentiate the functionality of CAR T cells. Contrary to our hypothesis, VSVmIFNß infection is associated with attrition of murine EGFRvIII CAR T cells in a B16EGFRvIII model, despite inducing a robust proinflammatory shift in the chemokine profile. Mechanistically, type I interferon (IFN) expressed following infection promotes apoptosis, activation, and inhibitory receptor expression, and interferon-insensitive CAR T cells enable combinatorial therapy with VSVmIFNß. Our study uncovers an unexpected mechanism of therapeutic interference, and prompts further investigation into the interaction between CAR T cells and oncolytic viruses to optimize combination therapy.


Subject(s)
Immunotherapy, Adoptive , Interferon-beta/metabolism , Oncolytic Viruses/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Animals , Apoptosis , Cell Line, Tumor , Chemokines/metabolism , Combined Modality Therapy , Female , Interferon-beta/genetics , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Receptors, Antigen, T-Cell/metabolism , Spleen/immunology
9.
Neuro Oncol ; 22(12): 1757-1770, 2020 12 18.
Article in English | MEDLINE | ID: mdl-32459315

ABSTRACT

BACKGROUND: Diffuse midline glioma, formerly DIPG (diffuse intrinsic pontine glioma), is the deadliest pediatric brainstem tumor with median survival of less than one year. Here, we investigated (i) whether direct delivery of adenovirus-expressing cluster of differentiation (CD)40 ligand (Ad-CD40L) to brainstem tumors would induce immune-mediated tumor clearance and (ii) if so, whether therapy would be associated with a manageable toxicity due to immune-mediated inflammation in the brainstem. METHODS: Syngeneic gliomas in the brainstems of immunocompetent mice were treated with Ad-CD40L and survival, toxicity, and immune profiles determined. A clinically translatable vector, whose replication would be tightly restricted to tumor cells, rAd-Δ24-CD40L, was tested in human patient-derived diffuse midline gliomas and immunocompetent models. RESULTS: Expression of Ad-CD40L restricted to brainstem gliomas by pre-infection induced complete rejection, associated with immune cell infiltration, of which CD4+ T cells were critical for therapy. Direct intratumoral injection of Ad-CD40L into established brainstem tumors improved survival and induced some complete cures but with some acute toxicity. RNA-sequencing analysis showed that Ad-CD40L therapy induced neuroinflammatory immune responses associated with interleukin (IL)-6, IL-1ß, and tumor necrosis factor α. Therefore, to generate a vector whose replication, and transgene expression, would be tightly restricted to tumor cells, we constructed rAd-Δ24-CD40L, the backbone of which has already entered clinical trials for diffuse midline gliomas. Direct intratumoral injection of rAd-Δ24-CD40L, with systemic blockade of IL-6 and IL-1ß, generated significant numbers of cures with readily manageable toxicity. CONCLUSIONS: Virus-mediated delivery of CD40L has the potential to be effective in treating diffuse midline gliomas without obligatory neuroinflammation-associated toxicity.


Subject(s)
Brain Stem Neoplasms , Glioma , Adenoviridae , Animals , Brain Stem Neoplasms/therapy , CD4-Positive T-Lymphocytes , CD40 Ligand , Glioma/therapy , Humans , Mice
10.
Nat Commun ; 11(1): 790, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034147

ABSTRACT

APOBEC3B, an anti-viral cytidine deaminase which induces DNA mutations, has been implicated as a mediator of cancer evolution and therapeutic resistance. Mutational plasticity also drives generation of neoepitopes, which prime anti-tumor T cells. Here, we show that overexpression of APOBEC3B in tumors increases resistance to chemotherapy, but simultaneously heightens sensitivity to immune checkpoint blockade in a murine model of melanoma. However, in the vaccine setting, APOBEC3B-mediated mutations reproducibly generate heteroclitic neoepitopes in vaccine cells which activate de novo T cell responses. These cross react against parental, unmodified tumors and lead to a high rate of cures in both subcutaneous and intra-cranial tumor models. Heteroclitic Epitope Activated Therapy (HEAT) dispenses with the need to identify patient specific neoepitopes and tumor reactive T cells ex vivo. Thus, actively driving a high mutational load in tumor cell vaccines increases their immunogenicity to drive anti-tumor therapy in combination with immune checkpoint blockade.


Subject(s)
Cancer Vaccines/pharmacology , Cytidine Deaminase/immunology , Immunotherapy/methods , Minor Histocompatibility Antigens/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Drug Resistance, Neoplasm , Epitopes/immunology , Female , Humans , Killer Cells, Natural/immunology , Melanoma/immunology , Melanoma/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Mutation , Tumor Escape/drug effects
11.
Mol Ther Oncolytics ; 16: 63-74, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-31930167

ABSTRACT

Genetically modified vesicular stomatitis virus (VSV) is an attractive agent for cancer treatment due to rapid intratumoral replication and observed clinical responses. Although VSV selectively kills malignant cells and can boost antitumor immunity, limited induction of intratumoral immune infiltration remains a barrier to efficacy in some cancer models. Here we engineered the oncolytic VSV platform to encode the T cell chemokine CXCL9, which is known to mediate the recruitment of activated CD8+ cytotoxic T cells and CD4+ T helper cells, and demonstrates conserved protein function between mice and humans. Chemotactic activity of the virally encoded chemokine was confirmed in vitro. Intratumoral concentration of CXCL9 was shown to increase after VSV therapy in three different cancer models, but to a much greater degree after VSV-CXCL9 therapy as compared with VSV control viruses. Despite a steep chemokine gradient from the tumor to the bloodstream, tumor trafficking of adoptively transferred and endogenous T cells was not measurably increased following VSV-CXCL9 therapy. Our results indicate that oncolytic VSV infection promotes release of CXCL9 in the tumor microenvironment, but further boosting of the functional chemokine gradient through virus engineering has little incremental impact on intratumoral immune cell infiltration in mouse and human tumor models.

12.
Stem Cell Reports ; 13(2): 307-321, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31378674

ABSTRACT

Generation of functional ß cells from pluripotent sources would accelerate diagnostic and therapeutic applications for diabetes research and therapy. However, it has been challenging to generate competent ß cells with dynamic insulin-secretory capacity to glucose and incretin stimulations. We introduced transcription factors, critical for ß-cell development and function, in differentiating human induced pluripotent stem cells (PSCs) and assessed the impact on the functionality of derived ß-cell (psBC) progeny. A perifusion system revealed stepwise transduction of the PDX1, NEUROG3, and MAFA triad (PNM) enabled in vitro generation of psBCs with glucose and GLP-1 responsiveness within 3 weeks. PNM transduction upregulated genes associated with glucose sensing, insulin secretion, and ß-cell maturation. In recipient diabetic mice, PNM-transduced psBCs showed glucose-responsive insulin secretion as early as 1 week post transplantation. Thus, enhanced pre-emptive ß-cell specification of PSCs by PNM drives generation of glucose- and incretin-responsive psBCs in vitro, offering a competent tissue-primed biotherapy.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Glucagon-Like Peptide 1/pharmacology , Glucose/pharmacology , Induced Pluripotent Stem Cells/transplantation , Insulin Secretion/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , C-Peptide/metabolism , Cell Differentiation , Diabetes Mellitus, Experimental/chemically induced , Gene Expression Regulation , Glucose Tolerance Test , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Mice, SCID , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transduction, Genetic , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Biochem Biophys Rep ; 17: 108-113, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30623114

ABSTRACT

Hyperinsulinemic hypoglycemia subtype glucokinase (GCK-HH) is caused by an activating mutation in glucokinase (GCK) and has been shown to increase ß-cell death. However, the mechanism of ß-cell death in GCK-HH remains poorly understood. Here, we expressed the GCK-HH V91L GCK mutant in INS-1 832/13 cells to determine the effect of the mutation on ß-cell viability and the mechanisms of ß-cell death. We showed that expression of the V91L GCK mutant in INS-1 832/13 cells resulted in a rapid glucose concentration-dependent loss of cell viability. At 11 mM D-glucose, INS-1 832/13 cells expressing V91L GCK showed increased cell permeability without significant increases in Annexin V staining or caspase 3/7 activation, indicating that these cells are primarily undergoing cell death via necrosis. Over-expression of SV40 large T antigen, which inhibits the p53 pathway, did not affect the V91L GCK-induced cell death. We also found that non-phosphorylatable L-glucose did not induce rapid cell death. Of note, glucose phosphorylation coincided with a 90% loss of intracellular ATP content. Thus, our data suggest that the GCK V91L mutant induces rapid necrosis in INS-1 cells through accelerated glucose phosphorylation, ATP depletion, and increased cell permeability.

14.
Dis Model Mech ; 11(6)2018 06 13.
Article in English | MEDLINE | ID: mdl-29915142

ABSTRACT

High-fat diet (HFD)-fed mouse models have been widely used to study early type 2 diabetes. Decreased ß-cell glucokinase (GCK) expression has been observed in HFD-induced diabetes. However, owing to its crucial roles in glucose metabolism in the liver and in islet ß-cells, the contribution of decreased GCK expression to the development of HFD-induced diabetes is unclear. Here, we employed a ß-cell-targeted gene transfer vector and determined the impact of ß-cell-specific increase in GCK expression on ß-cell function and glucose handling in vitro and in vivo Overexpression of GCK enhanced glycolytic flux, ATP-sensitive potassium channel activation and membrane depolarization, and increased proliferation in Min6 cells. ß-cell-targeted GCK transduction did not change glucose handling in chow-fed C57BL/6 mice. Although adult mice fed a HFD showed reduced islet GCK expression, impaired glucose tolerance and decreased glucose-stimulated insulin secretion (GSIS), ß-cell-targeted GCK transduction improved glucose tolerance and restored GSIS. Islet perifusion experiments verified restored GSIS in isolated HFD islets by GCK transduction. Thus, our data identify impaired ß-cell GCK expression as an underlying mechanism for dysregulated ß-cell function and glycemic control in HFD-induced diabetes. Our data also imply an etiological role of GCK in diet-induced diabetes.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Glucokinase/metabolism , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Animals , Calcium/metabolism , Cell Proliferation , Dependovirus/metabolism , Diabetes Mellitus, Experimental/genetics , Diet, High-Fat , Glucose/metabolism , Glucose Tolerance Test , Glycolysis , Insulin/metabolism , Intracellular Space/metabolism , Male , Mice, Inbred C57BL , Signal Transduction , Transduction, Genetic , Up-Regulation/genetics
15.
BMC Res Notes ; 10(1): 720, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29221488

ABSTRACT

OBJECTIVE: Recombinant adeno-associated virus (AAV)-based vectors are characterized by their robust and safe transgene delivery. The CRISPR/Cas9 and guide RNA (gRNA) system present a promising genome-editing platform, and a recent development of a shorter Cas9 enzyme from Staphylococcus aureus (SaCas9) allows generation of high titer single AAV vectors which carry both saCas9- and gRNA-expression cassettes. Here, we used two AAV-SaCas9 vectors with distinct GFP-targeted gRNA sequences and determined the impact of AAV-SaCas9-gRNA vector treatment in a single cell clone carrying a GFP-expression cassette. RESULTS: Our results showed comparable GFP knockout efficiencies (40-50%) upon a single low-dose infection. Three consecutive transductions of 25-fold higher doses of vectors showed 80% GFP knockout efficiency. To analyze the "AAV-SaCas9-resistant cell population", we sorted the residual GFP-positive cells and assessed their permissiveness to super-infection with two AAV-Cas9-GFP vectors. We found the sorted cells were significantly more resistant to the GFP knockout mediated by the same AAV vector, but not by the other GFP-targeted AAV vector. Our data therefore demonstrate highly efficient genome-editing by the AAV-SaCas9-gRNA vector system. Differential susceptibilities of single cell-derived cells to the AAV-SaCas9-gRNA-mediated genome editing may represent a formidable barrier to achieve 100% genome editing efficiency by this vector system.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Dependovirus , Endonucleases/genetics , Exome Sequencing , Gene Editing , Genetic Vectors , RNA, Guide, Kinetoplastida , Staphylococcus aureus/enzymology , Bacterial Proteins , CRISPR-Associated Protein 9 , Cell Line , Disease Susceptibility , Green Fluorescent Proteins , HEK293 Cells , Humans
16.
Stem Cells Transl Med ; 5(5): 694-702, 2016 May.
Article in English | MEDLINE | ID: mdl-26987352

ABSTRACT

UNLABELLED: Human induced pluripotent stem cells (iPSCs) and derived progeny provide invaluable regenerative platforms, yet their clinical translation has been compromised by their biosafety concern. Here, we assessed the safety of transplanting patient-derived iPSC-generated pancreatic endoderm/progenitor cells. Transplantation of progenitors from iPSCs reprogrammed by lentiviral vectors (LV-iPSCs) led to the formation of invasive teratocarcinoma-like tumors in more than 90% of immunodeficient mice. Moreover, removal of primary tumors from LV-iPSC progeny-transplanted hosts generated secondary and metastatic tumors. Combined transgene-free (TGF) reprogramming and elimination of residual pluripotent cells by enzymatic dissociation ensured tumor-free transplantation, ultimately enabling regeneration of type 1 diabetes-specific human islet structures in vivo. The incidence of tumor formation in TGF-iPSCs was titratable, depending on the oncogenic load, with reintegration of the cMYC expressing vector abolishing tumor-free transplantation. Thus, transgene-free cMYC-independent reprogramming and elimination of residual pluripotent cells are mandatory steps in achieving transplantation of iPSC progeny for customized and safe islet regeneration in vivo. SIGNIFICANCE: Pluripotent stem cell therapy for diabetes relies on the safety as well as the quality of derived insulin-producing cells. Data from this study highlight prominent tumorigenic risks of induced pluripotent stem cell (iPSC) products, especially when reprogrammed with integrating vectors. Two major underlying mechanisms in iPSC tumorigenicity are residual pluripotent cells and cMYC overload by vector integration. This study also demonstrated that combined transgene-free reprogramming and enzymatic dissociation allows teratoma-free transplantation of iPSC progeny in the mouse model in testing the tumorigenicity of iPSC products. Further safety assessment and improvement in iPSC specification into a mature ß cell phenotype would lead to safe islet replacement therapy for diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Diabetes Mellitus, Type 2/surgery , Induced Pluripotent Stem Cells/transplantation , Islets of Langerhans Transplantation/methods , Islets of Langerhans/surgery , Keratinocytes/transplantation , Regeneration , Teratocarcinoma/prevention & control , Adult , Aged , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming Techniques , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Gene Expression Regulation, Neoplastic , Genetic Vectors , Heterografts , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/adverse effects , Keratinocytes/metabolism , Keratinocytes/pathology , Lentivirus/genetics , Male , Mice, SCID , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Transfection
17.
Diabetes ; 65(6): 1606-15, 2016 06.
Article in English | MEDLINE | ID: mdl-26983960

ABSTRACT

Considerable evidence implicates cellular senescence in the biology of aging and chronic disease. Diet and exercise are determinants of healthy aging; however, the extent to which they affect the behavior and accretion of senescent cells within distinct tissues is not clear. Here we tested the hypothesis that exercise prevents premature senescent cell accumulation and systemic metabolic dysfunction induced by a fast-food diet (FFD). Using transgenic mice that express EGFP in response to activation of the senescence-associated p16(INK4a) promoter, we demonstrate that FFD consumption causes deleterious changes in body weight and composition as well as in measures of physical, cardiac, and metabolic health. The harmful effects of the FFD were associated with dramatic increases in several markers of senescence, including p16, EGFP, senescence-associated ß-galactosidase, and the senescence-associated secretory phenotype (SASP) specifically in visceral adipose tissue. We show that exercise prevents the accumulation of senescent cells and the expression of the SASP while nullifying the damaging effects of the FFD on parameters of health. We also demonstrate that exercise initiated after long-term FFD feeding reduces senescent phenotype markers in visceral adipose tissue while attenuating physical impairments, suggesting that exercise may provide restorative benefit by mitigating accrued senescent burden. These findings highlight a novel mechanism by which exercise mediates its beneficial effects and reinforces the effect of modifiable lifestyle choices on health span.


Subject(s)
Adipose Tissue/cytology , Cellular Senescence/physiology , Diet/adverse effects , Fast Foods/adverse effects , Physical Conditioning, Animal/physiology , Aging/physiology , Animals , Body Composition , Body Weight , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , beta-Galactosidase/metabolism
18.
Hypertension ; 66(1): 199-210, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26063669

ABSTRACT

Altered myocardial structure and function, secondary to chronically elevated blood pressure, are leading causes of heart failure and death. B-type natriuretic peptide (BNP), a guanylyl cyclase A agonist, is a cardiac hormone integral to cardiovascular regulation. Studies have demonstrated a causal relationship between reduced production or impaired BNP release and the development of human hypertension. However, the consequences of BNP insufficiency on blood pressure and hypertension-associated complications remain poorly understood. Therefore, the goal of this study was to create and characterize a novel model of BNP deficiency to investigate the effects of BNP absence on cardiac and renal structure, function, and survival. Genetic BNP deletion was generated in Dahl salt-sensitive rats. Compared with age-matched controls, BNP knockout rats demonstrated adult-onset hypertension. Increased left ventricular mass with hypertrophy and substantially augmented hypertrophy signaling pathway genes, developed in young adult knockout rats, which preceded hypertension. Prolonged hypertension led to increased cardiac stiffness, cardiac fibrosis, and thrombi formation. Significant elongation of the QT interval was detected at 9 months in knockout rats. Progressive nephropathy was also noted with proteinuria, fibrosis, and glomerular alterations in BNP knockout rats. End-organ damage contributed to a significant decline in overall survival. Systemic BNP overexpression reversed the phenotype of genetic BNP deletion. Our results demonstrate the critical role of BNP defect in the development of systemic hypertension and associated end-organ damage in adulthood.


Subject(s)
Disease Models, Animal , Hypertension/etiology , Natriuretic Peptide, Brain/physiology , Age of Onset , Animals , Compliance , Death, Sudden, Cardiac/etiology , Fibrosis , Gene Expression Regulation , Gene Knockout Techniques , Humans , Hypertension/genetics , Hypertension/pathology , Hypertension/prevention & control , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/pathology , Kidney Glomerulus/pathology , Long QT Syndrome/etiology , Myocardial Contraction/genetics , Myocardium/pathology , Natriuretic Peptide, Brain/deficiency , Natriuretic Peptide, Brain/genetics , Phenotype , Rats , Rats, Inbred Dahl , Recombinant Fusion Proteins/metabolism , Renal Insufficiency, Chronic/etiology , Signal Transduction/genetics , Signal Transduction/physiology
19.
Stem Cell Res Ther ; 6: 48, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25889591

ABSTRACT

INTRODUCTION: Advances in the field of stem cells have led to novel avenues for generating induced pluripotent stem cells (iPSCs) from differentiated somatic cells. iPSCs are typically obtained by the introduction of four factors--OCT4, SOX2, KLF4, and cMYC--via integrating vectors. Here, we report the feasibility of a novel reprogramming process based on vectors derived from the non-integrating vaccine strain of measles virus (MV). METHODS: We produced a one-cycle MV vector by substituting the viral attachment protein gene with the green fluorescent protein (GFP) gene. This vector was further engineered to encode for OCT4 in an additional transcription unit. RESULTS: After verification of OCT4 expression, we assessed the ability of iPSC reprogramming. The reprogramming vector cocktail with the OCT4-expressing MV vector and SOX2-, KLF4-, and cMYC-expressing lentiviral vectors efficiently transduced human skin fibroblasts and formed iPSC colonies. Reverse transcription-polymerase chain reaction and immunostaining confirmed induction of endogenous pluripotency-associated marker genes, such as SSEA-4, TRA-1-60, and Nanog. Pluripotency of derived clones was confirmed by spontaneous differentiation into three germ layers, teratoma formation, and guided differentiation into beating cardiomyocytes. CONCLUSIONS: MV vectors can induce efficient nuclear reprogramming. Given the excellent safety record of MV vaccines and the translational capabilities recently developed to produce MV-based vectors now used for cancer clinical trials, our MV vector system provides an RNA-based, non-integrating gene transfer platform for nuclear reprogramming that is amenable for immediate clinical translation.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Measles virus/genetics , Myocytes, Cardiac/cytology , Octamer Transcription Factor-3/genetics , Animals , Antigens, Surface/genetics , Biomarkers , Cell Line , Cellular Reprogramming/physiology , Chlorocebus aethiops , Fibroblasts/cytology , Foreskin/cytology , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, SCID , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Proteoglycans/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , Skin/cytology , Stage-Specific Embryonic Antigens/genetics , Vero Cells
20.
Diabetologia ; 58(2): 313-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25338552

ABSTRACT

AIMS/HYPOTHESIS: Achieving a better understanding of beta cell regeneration after immunological destruction is crucial for the development of immunotherapy approaches for type 1 diabetes. In previous type 1 diabetes models, sustained immune activation eliminates regenerating beta cells, thus limiting the study of the regenerative capacity of beta cells upon immunological destruction. Here, we employed an adeno-associated virus 8 (AAV8) vector for beta cell-targeted overexpression of a foreign antigen to induce single-round immunological destruction of existing beta cells. METHODS: Young and aged C57BL/6J mice were treated with AAV8 vectors expressing the foreign antigen luciferase. Islet inflammation and regeneration was observed at 3, 6, 10 and 22 weeks post-AAV delivery. RESULTS: In young C57BL/6J mice, robust humoral and cellular immune responses were developed towards antigen-expressing beta cells, leading to decreased beta cell mass. This was followed by beta cell mass replenishment, along with enhanced proliferation of insulin-positive cells, recruitment of nestin/CD34-positive endothelial cells, displacement of alpha cells and mobilisation of cytoplasmic neurogenin 3-positive cells. Mice with recovering beta cells showed normal or reduced fasting blood glucose levels and faster glucose clearance than controls. Although aged mice demonstrated similar responses to the treatment, they initially exhibited notable islet scarring and fluctuations in blood glucose levels, indicating that beta cell regeneration is slower in aged mice. CONCLUSIONS/INTERPRETATION: Our hit-and-run, beta cell-targeted antigen expression system provides an opportunity to monitor the impact of single-round immunological beta cell destruction in animals with diverse genetic backgrounds or ageing status.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Regeneration/immunology , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Dependovirus , Disease Models, Animal , Genetic Vectors , Immunosuppressive Agents/pharmacology , Insulin-Secreting Cells/immunology , Male , Mice , Mice, Inbred C57BL , Parvoviridae Infections , Regeneration/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL