Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
Mol Ther Nucleic Acids ; 35(3): 102282, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39176174

ABSTRACT

Although our understanding of herpes simplex virus type 1 (HSV-1) biology has been considerably enhanced, developing therapeutic strategies to eliminate HSV-1 in latently infected individuals remains a public health concern. Current antiviral drugs used for the treatment of HSV-1 complications are not specific and do not address latent infection. We recently developed a CRISPR-Cas9-based gene editing platform to specifically target the HSV-1 genome. In this study, we further used 2D Vero cell culture and 3D human induced pluripotent stem cell-derived cerebral organoid (CO) models to assess the effectiveness of our editing constructs targeting viral ICP0 or ICP27 genes. We found that targeting the ICP0 or ICP27 genes with AAV2-CRISPR-Cas9 vectors in Vero cells drastically suppressed HSV-1 replication. In addition, we productively infected COs with HSV-1, characterized the viral replication kinetics, and established a viral latency model. Finally, we discovered that ICP0- or ICP27-targeting AAV2-CRISPR-Cas9 vector significantly reduced viral rebound in the COs that were latently infected with HSV-1. In summary, our results suggest that CRISPR-Cas9 gene editing of HSV-1 is an efficient therapeutic approach to eliminate the latent viral reservoir and treat HSV-1-associated complications.

2.
J Neurovirol ; 30(1): 71-85, 2024 02.
Article in English | MEDLINE | ID: mdl-38355914

ABSTRACT

Mixed glia are infiltrated with HIV-1 virus early in the course of infection leading to the development of a persistent viral reservoir in the central nervous system. Modification of the HIV-1 genome using gene editing techniques, including CRISPR/Cas9, has shown great promise towards eliminating HIV-1 viral reservoirs; whether these techniques are capable of removing HIV-1 viral proteins from mixed glia, however, has not been systematically evaluated. Herein, the efficacy of adeno-associated virus 9 (AAV9)-CRISPR/Cas9 gene editing for eliminating HIV-1 messenger RNA (mRNA) from cortical mixed glia was evaluated in vitro and in vivo. In vitro, a within-subjects experimental design was utilized to treat mixed glia isolated from neonatal HIV-1 transgenic (Tg) rats with varying doses (0, 0.9, 1.8, 2.7, 3.6, 4.5, or 5.4 µL corresponding to a physical titer of 0, 4.23 × 109, 8.46 × 109, 1.269 × 1010, 1.692 × 1010, 2.115 × 1010, and 2.538 × 1010 gc/µL) of CRISPR/Cas9 for 72 h. Dose-dependent decreases in the number of HIV-1 mRNA, quantified using an innovative in situ hybridization technique, were observed in a subset (i.e., n = 5 out of 8) of primary mixed glia. In vivo, HIV-1 Tg rats were retro-orbitally inoculated with CRISPR/Cas9 for two weeks, whereby treatment resulted in profound excision (i.e., approximately 53.2%) of HIV-1 mRNA from the medial prefrontal cortex. Given incomplete excision of the HIV-1 viral genome, the clinical relevance of HIV-1 mRNA knockdown for eliminating neurocognitive impairments was evaluated via examination of temporal processing, a putative neurobehavioral mechanism underlying HIV-1-associated neurocognitive disorders (HAND). Indeed, treatment with CRISPR/Cas9 protractedly, albeit not permanently, restored the developmental trajectory of temporal processing. Proof-of-concept studies, therefore, support the susceptibility of mixed glia to gene editing and the potential of CRISPR/Cas9 to serve as a novel therapeutic strategy for HAND, even in the absence of full viral eradication.


Subject(s)
CRISPR-Cas Systems , Gene Editing , HIV-1 , RNA, Messenger , Rats, Transgenic , Animals , HIV-1/genetics , HIV-1/physiology , Rats , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Editing/methods , Neuroglia/virology , Neuroglia/metabolism , Dependovirus/genetics , HIV Infections/virology , HIV Infections/genetics , Gene Knockdown Techniques , RNA, Viral/genetics , Cognition/physiology , Humans
4.
Res Sq ; 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37886577

ABSTRACT

Mixed glia are infiltrated with HIV-1 virus early in the course of infection leading to the development of a persistent viral reservoir in the central nervous system. Modification of the HIV-1 genome using gene editing techniques, including CRISPR/Cas9, has shown great promise towards eliminating HIV-1 viral reservoirs; whether these techniques are capable of removing HIV-1 viral proteins from mixed glia, however, has not been systematically evaluated. Herein, the efficacy of adeno-associated virus 9 (AAV9)-CRISPR/Cas9 gene editing for eliminating HIV-1 mRNA from cortical mixed glia was evaluated in vitro and in vivo. In vitro, a within-subjects experimental design was utilized to treat mixed glia isolated from neonatal HIV-1 transgenic (Tg) rats with varying doses (0, 0.9, 1.8, 2.7, 3.6, 4.5, or 5.4 µL) of CRISPR/Cas9 for 72 hours. Dose-dependent decreases in the number of HIV-1 mRNA, quantified using an innovative in situ hybridization technique, were observed in a subset (i.e., n=5 out of 8) of primary mixed glia. In vivo, HIV-1 Tg rats were retro-orbitally inoculated with CRISPR/Cas9 for two weeks, whereby treatment resulted in profound excision (i.e., approximately 53.2%) of HIV-1 mRNA from the mPFC. Given incomplete excision of the HIV-1 viral genome, the clinical relevance of HIV-1 mRNA knockdown for eliminating neurocognitive impairments was evaluated via examination of temporal processing, a putative neurobehavioral mechanism underlying HIV-1 associated neurocognitive disorders (HAND). Indeed, treatment with CRISPR/Cas9 partially restored the developmental trajectory of temporal processing. Proof-of-concept studies, therefore, support the susceptibility of mixed glia to gene editing and the potential of CRISPR/Cas9 to serve as a novel therapeutic strategy for HAND, even in the absence of full viral eradication.

5.
Nanomedicine (Lond) ; 18(20): 1343-1360, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37815117

ABSTRACT

Aim: To develop lipid nano-antiretrovirals (LNAs) for the treatment of HIV-infected macrophages. Materials & methods: LNAs were prepared with docosahexaenoic acid to facilitate brain penetration and surface-decorated with folate considering that infected macrophages often overexpress folate receptors. Results: Folate-decorated LNAs loading rilpivirine (RPV) were efficiently taken up by folate receptor-expressing cell types including activated macrophages. The intracellular Cmax of the RPV-LNAs in activated macrophages was 2.54-fold and the area under the curve was 3.4-fold versus free RPV, translating to comparable or higher (p < 0.01; RPV ≤6.5 ng/ml) activities against HIV infectivity and superior protection (p < 0.05) against HIV cytotoxicity. LNAs were also effective in monocyte-derived macrophages. Conclusion: These findings demonstrate the potential of LNAs for the treatment of infected macrophages, which are key players in HIV reservoirs.


HIV can infect and hide inside certain types of white blood cells that make up the immune system and help defend our body, such as macrophages. Because these infected cells tend to carry the virus to certain organs where antiviral drugs have a hard time reaching, the virus is able to avoid treatment from the drug. In this study, the authors developed very small devices known as nanocarriers to carry antiviral drugs. These nanocarriers were designed to seek out infected macrophages. The nanocarriers were successfully built with oils and lipids that are safe for patients and could easily deliver antiviral drugs to macrophages infected by HIV. Excellent anti-HIV effects were observed using these nanocarriers. In summary, the authors developed a promising device with the potential to fight HIV in a smart and safe manner.


Subject(s)
Anti-HIV Agents , HIV Infections , Humans , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Rilpivirine/therapeutic use , Anti-Retroviral Agents , Macrophages , Lipids/therapeutic use , Folic Acid
6.
JACC Basic Transl Sci ; 8(7): 820-839, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37547075

ABSTRACT

B-cell lymphoma 2-associated athanogene-3 (Bag3) is expressed in all animal species, with Bag3 levels being most prominent in the heart, the skeletal muscle, the central nervous system, and in many cancers. Preclinical studies of Bag3 biology have focused on animals that have developed compromised cardiac function; however, the present studies were performed to identify the pathways perturbed in the heart even before the occurrence of clinical signs of dilatation and failure of the heart. These studies show that hearts carrying variants that knockout one allele of BAG3 have significant alterations in multiple cellular pathways including apoptosis, autophagy, mitochondrial homeostasis, and the inflammasome.

7.
Gene Ther ; 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37587230

ABSTRACT

In this study, we demonstrate the safety and utility of CRISPR-Cas9 gene editing technology for in vivo editing of proviral DNA in ART-treated, virally controlled simian immunodeficiency virus (SIV) infected rhesus macaques, an established model for HIV infection. EBT-001 is an AAV9-based vector delivering SaCas9 and dual guide RNAs designed to target multiple regions of the SIV genome: the viral LTRs, and the Gag gene. The results presented here demonstrate that a single IV inoculation of EBT-001 at each of 3 dose levels (1.4 × 1012, 1.4 × 1013 and 1.4 × 1014 genome copies/kg) resulted in broad and functional biodistribution of AAV9-EBT-001 to known tissue reservoirs of SIV. No off-target effects or abnormal pathology were observed, and animals returned to their normal body weight after receiving EBT-001. Importantly, the macaques that received the 2 highest doses of EBT-001 showed improved absolute lymphocyte counts as compared to antiretroviral-treated controls. Taken together, these results demonstrate safety, biodistribution, and in vivo proviral DNA editing following IV administration of EBT-001, supporting the further development of CRISPR-based gene editing as a potential therapeutic approach for HIV in humans.

8.
Mol Ther Nucleic Acids ; 32: 1010-1025, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37346975

ABSTRACT

Post-translational glycosylation of the HIV-1 envelope protein involving precursor glycan trimming by mannosyl oligosaccharide glucosidase (MOGS) is critically important for morphogenesis of virions and viral entry. Strategic editing of the MOGS gene in T lymphocytes and myeloid origin cells harboring latent proviral DNA results in the production of non-infectious particles upon treatment of cells with latency reversal agents. Controlled activation of CRISPR-MOGS by rebound HIV-1 mitigates production of infectious particles that exhibit poor ability of the virus to penetrate uninfected cells. Moreover, exclusive activation of CRISPR in cells infected with HIV-1 alleviates concern for broad off-target impact of MOGS gene ablation in uninfected cells. Combination CRISPR treatment of peripheral blood lymphocytes prepared from blood of people with HIV-1 (PWH) tailored for editing the MOGS gene (CRISPR-MOGS) and proviral HIV-1 DNA (CRISPR-HIV) revealed a cooperative impact of CRISPR treatment in inhibiting the production of infectious HIV-1 particles. Our design for genetic inactivation of MOGS by CRISPR exhibits no detectable off-target effects on host cells or any deleterious impact on cell survival and proliferation. Our findings offer the development of a new combined gene editing-based cure strategy for the diminution of HIV-1 spread after cessation of antiretroviral therapy (ART) and its elimination.

9.
Proc Natl Acad Sci U S A ; 120(19): e2217887120, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37126704

ABSTRACT

Treatment of HIV-1ADA-infected CD34+ NSG-humanized mice with long-acting ester prodrugs of cabotegravir, lamivudine, and abacavir in combination with native rilpivirine was followed by dual CRISPR-Cas9 C-C chemokine receptor type five (CCR5) and HIV-1 proviral DNA gene editing. This led to sequential viral suppression, restoration of absolute human CD4+ T cell numbers, then elimination of replication-competent virus in 58% of infected mice. Dual CRISPR therapies enabled the excision of integrated proviral DNA in infected human cells contained within live infected animals. Highly sensitive nucleic acid nested and droplet digital PCR, RNAscope, and viral outgrowth assays affirmed viral elimination. HIV-1 was not detected in the blood, spleen, lung, kidney, liver, gut, bone marrow, and brain of virus-free animals. Progeny virus from adoptively transferred and CRISPR-treated virus-free mice was neither detected nor recovered. Residual HIV-1 DNA fragments were easily seen in untreated and viral-rebounded animals. No evidence of off-target toxicities was recorded in any of the treated animals. Importantly, the dual CRISPR therapy demonstrated statistically significant improvements in HIV-1 cure percentages compared to single treatments. Taken together, these observations underscore a pivotal role of combinatorial CRISPR gene editing in achieving the elimination of HIV-1 infection.


Subject(s)
HIV Infections , HIV Seropositivity , Mice , Animals , Humans , Anti-Retroviral Agents/therapeutic use , Gene Editing , Proviruses/genetics , Receptors, CCR5
10.
Int J Mol Sci ; 23(17)2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36077131

ABSTRACT

Protein quality control allows eukaryotes to maintain proteostasis under the stress of constantly changing conditions. In this review, we discuss the current literature on PQC, highlighting flaws that must exist for malignancy to occur. At the nidus of PQC, the expression of BAG1-6 reflects the cell environment; each isoform directs proteins toward different, parallel branches of the quality control cascade. The sum of these branches creates a net shift toward either homeostasis or apoptosis. With an established role in ALP, Bag3 is necessary for cell survival in stress conditions including those of the cancerous niche (i.e., hypoxia, hypermutation). Evidence suggests that excessive Bag3-HSP70 activity not only sustains, but also propagates cancers. Its role is anti-apoptotic-which allows malignant cells to persist-and intercellular-with the production of infectious 'oncosomes' enabling cancer expansion and recurrence. While Bag3 has been identified as a key prognostic indicator in several cancer types, its investigation is limited regarding glioblastoma. The cochaperone HSP70 has been strongly linked with GBM, while ALP inhibitors have been shown to improve GBM susceptibility to chemotherapeutics. Given the highly resilient, frequently recurrent nature of GBM, the targeting of Bag3 is a necessary consideration for the successful and definitive treatment of GBM.


Subject(s)
Glioblastoma , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy , Glioblastoma/drug therapy , HSP70 Heat-Shock Proteins/metabolism , Humans , Transcription Factors/metabolism
11.
Methods Mol Biol ; 2311: 51-61, 2021.
Article in English | MEDLINE | ID: mdl-34033077

ABSTRACT

The research on human neural progenitor cells holds great potential for the understanding of the molecular programs that control differentiation of cells of glial and neuronal lineages, as well as pathogenetic mechanisms of neurological diseases. Stem cell technologies also provide opportunities for the pharmaceutical industry to develop new approaches for regenerative medicine. Here, we describe the protocol for the isolation and maintenance of neural progenitor cells and cortical neurons using human fetal brain tissue. This protocol can be successfully adapted for the preparation of rodent neural and oligodendrocyte progenitor cells. While several methods for isolating neural and oligodendrocyte progenitors from rodent brain tissue have been described, including techniques utilizing gene transfer and magnetic resonance beads, few methods are specifically focused on deriving human oligodendrocyte progenitor cells. Development of the human cultures provides the most physiologically relevant system for investigating mechanisms which regulate the function of oligodendrocytes, specifically of human origin.


Subject(s)
Cell Separation , Cerebral Cortex/physiology , Embryonic Stem Cells/physiology , Neural Stem Cells/physiology , Neurogenesis , Neurons/physiology , Primary Cell Culture , Animals , Cell Lineage , Cell Proliferation , Cells, Cultured , Cerebral Cortex/embryology , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Gestational Age , Humans , Neural Stem Cells/metabolism , Phenotype , Pregnancy , Rats , Rats, Sprague-Dawley
12.
Methods Mol Biol ; 2311: 161-166, 2021.
Article in English | MEDLINE | ID: mdl-34033084

ABSTRACT

The culturing of neurons results in formation of the layer of neurons with random extensive overlapping outgrowth. To understand specific roles of somas, axons, and dendrites in complex function of neurons and to identify molecular mechanisms of biological processes in these cellular compartments, various methods were developed. We utilized AXon Investigation System (AXIS™) manufactured by Millipore. This device provides an opportunity to orient neuronal outgrowth and spatially isolate neuronal processes from neuronal bodies. AXIS device is a slide-mounted microfluidic system, which consists of four wells. Two of the wells are connected by a channel on each side of the device. Channels are connected by microgrooves (approximately 120). The size of microgrooves (10µm in width and 5µm in height) does not permit passage of cell through while allowing extension of neurites. The microfluidic design also allows for an establishment of a hydrostatic gradient when the volume in one chamber is greater than that in the other (Park et al., Nat Protoc 1:2128-2136, 2006). This feature allows for studying of the effect of specific compounds on selected compartments of neurons.


Subject(s)
Cell Culture Techniques/instrumentation , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Neuronal Outgrowth , Neurons/physiology , Cells, Cultured , Equipment Design , Fetus , Gestational Age , Humans , Hydrostatic Pressure , Neuronal Outgrowth/drug effects , Neurons/drug effects , Neurons/metabolism
13.
Methods Mol Biol ; 2311: 195-201, 2021.
Article in English | MEDLINE | ID: mdl-34033088

ABSTRACT

The lack of a convenient in vitro human neuronal model to study alcohol-induced neurodegenerative diseases, such as fetal alcohol syndrome (FAS), prompted us to develop human neuronal culture and in vitro human FAS model by incubating cells with physiologically relevant EtOH concentration (50 mM). Here, we describe the detailed method of isolation of human neuronal culture, and ability to analyze neurites extension using Sholl assay. We utilized highly efficient transfection method of neuronal cells to study morphology of neurons with or without EtOH treatment.


Subject(s)
Biological Assay , Brain/drug effects , Ethanol/toxicity , Fetal Alcohol Spectrum Disorders/pathology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Animals , Brain/embryology , Cell Separation , Cells, Cultured , Gestational Age , Humans , Neurons/pathology , Primary Cell Culture , Transfection
14.
Mol Cancer Res ; 19(8): 1375-1388, 2021 08.
Article in English | MEDLINE | ID: mdl-33863814

ABSTRACT

Asparagine synthetase (ASNS) is a gene on the long arm of chromosome 7 that is copy-number amplified in the majority of glioblastomas. ASNS copy-number amplification is associated with a significantly decreased survival. Using patient-derived glioma stem cells (GSC), we showed that significant metabolic alterations occur in gliomas when perturbing the expression of ASNS, which is not merely restricted to amino acid homeostasis. ASNS-high GSCs maintained a slower basal metabolic profile yet readily shifted to a greatly increased capacity for glycolysis and oxidative phosphorylation when needed. This led ASNS-high cells to a greater ability to proliferate and spread into brain tissue. Finally, we demonstrate that these changes confer resistance to cellular stress, notably oxidative stress, through adaptive redox homeostasis that led to radiotherapy resistance. Furthermore, ASNS overexpression led to modifications of the one-carbon metabolism to promote a more antioxidant tumor environment revealing a metabolic vulnerability that may be therapeutically exploited. IMPLICATIONS: This study reveals a new role for ASNS in metabolic control and redox homeostasis in glioma stem cells and proposes a new treatment strategy that attempts to exploit one vulnerable metabolic node within the larger multilayered tumor network.


Subject(s)
Asparagine/biosynthesis , Brain Stem Neoplasms/metabolism , Brain/metabolism , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Oxidative Stress/physiology , Animals , Aspartate-Ammonia Ligase/metabolism , HEK293 Cells , Humans , Mice , Retrospective Studies
15.
Cell Death Discov ; 7(1): 60, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33771978

ABSTRACT

Despite increasing numbers of aged individuals living with HIV, the mechanisms underlying HIV-associated neurological disorders (HANDs) remain elusive. As HIV-1 pathogenesis and aging are characterized by oxidative stress as well as altered protein quality control (PQC), reactive oxygen species (ROS) themselves might constitute a molecular mediator of neuronal PQC by modulating BCL-2 associated athanogene (BAG) family members. Present results reveal H2O2 replicated and exacerbated a reduction in neuronal BAG3 induced by the expression of HIV-1 viral proteins (i.e., Tat and Nef), while also causing an upregulation of BAG1. Such a reciprocal regulation of BAG3 and BAG1 levels was also indicated in two animal models of HIV, the doxycycline-inducible Tat (iTat) and the Tg26 mouse. Inhibiting oxidative stress via antioxidants in primary culture was capable of partially preserving neuronal BAG3 levels as well as electrophysiological functioning otherwise altered by HIV-1 viral proteins. Current findings indicate HIV-1 viral proteins and H2O2 may mediate neuronal PQC by exerting synergistic effects on complementary BAG family members, and suggest novel therapeutic targets for the aging HIV-1 population.

16.
Aging (Albany NY) ; 13(1): 1510-1527, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33472174

ABSTRACT

Despite a growing proportion of aged individuals at risk for developing cancer in the brain, the prognosis for these conditions remains abnormally poor due to limited knowledge of underlying mechanisms and minimal treatment options. While cancer metabolism in other organs is commonly associated with upregulated glycolysis (i.e. Warburg effect) and hyperactivation of PIK3/AKT/mTOR (PAM) pathways, the unique bioenergetic demands of the central nervous system may interact with these oncogenic processes to promote tumor progression in aging. Specifically, constitutive glycolysis and PIK3/AKT/mTOR signaling in glia may be dysregulated by age-dependent alterations in neurometabolic demands, ultimately contributing to pathological processes otherwise associated with PIK3/AKT/mTOR induction (e.g. cell cycle entry, impaired autophagy, dysregulated inflammation). Although several limitations to this theoretical model exist, the consideration of aberrant PIK3/AKT/mTOR signaling in glia during aging elucidates several therapeutic opportunities for brain tumors, including non-pharmacological interventions.


Subject(s)
Aging/metabolism , Brain Neoplasms/metabolism , Neuroglia/metabolism , Signal Transduction/physiology , Animals , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
17.
Viruses ; 13(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499370

ABSTRACT

JC Virus (JCPyV), a member of the Polyomaviridiæ family, is a human neurotropic virus with world-wide distribution. JCPyV is the established opportunistic infectious agent of progressive multifocal leukoencephalopathy, a fatal demyelinating disease, which results from the cytolytic infection of oligodendrocytes. Mutations in the regulatory region of JCPyV determine the different viral strains. Mad-1 the strain associated with PML contains two 98 base pair repeats, whereas the archetype strain (CY), which is the transmissible form of JCPyV, contains only one 98 tandem with two insertions of 62 and 23 base pairs respectively. The oncogenicity of JCPyV has been suspected since direct inoculation into the brain of rodents and primates resulted in the development of brain tumors and has been attributed to the viral protein, T-Antigen. To further understand the oncogenicity of JCPyV, a transgenic mouse colony containing the early region of the archetype strain (CY), under the regulation of its own promoter was generated. These transgenic animals developed tumors of neural crest origin, including: primitive neuroectodermal tumors, medulloblastomas, adrenal neuroblastomas, pituitary tumors, malignant peripheral nerve sheath tumors, and glioblastomas. Neoplastic cells from all different phenotypes express T-Antigen. The close parallels between the tumors developed by these transgenic animals and human CNS tumors make this animal model an excellent tool for the study of viral oncogenesis.


Subject(s)
Antigens, Viral, Tumor/physiology , Brain Neoplasms/virology , JC Virus/pathogenicity , Leukoencephalopathy, Progressive Multifocal/virology , Animals , Antigens, Viral, Tumor/genetics , Brain/metabolism , Brain Neoplasms/pathology , Capsid Proteins/genetics , Disease Models, Animal , Humans , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/pathology , Mice , Mice, Transgenic , Viral Proteins/genetics
18.
Nat Commun ; 11(1): 6065, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247091

ABSTRACT

Elimination of HIV DNA from infected individuals remains a challenge in medicine. Here, we demonstrate that intravenous inoculation of SIV-infected macaques, a well-accepted non-human primate model of HIV infection, with adeno-associated virus 9 (AAV9)-CRISPR/Cas9 gene editing construct designed for eliminating proviral SIV DNA, leads to broad distribution of editing molecules and precise cleavage and removal of fragments of the integrated proviral DNA from the genome of infected blood cells and tissues known to be viral reservoirs including lymph nodes, spleen, bone marrow, and brain among others. Accordingly, AAV9-CRISPR treatment results in a reduction in the percent of proviral DNA in blood and tissues. These proof-of-concept observations offer a promising step toward the elimination of HIV reservoirs in the clinic.


Subject(s)
Anti-Retroviral Agents/pharmacology , CRISPR-Cas Systems/genetics , DNA, Viral/genetics , Gene Editing , Proviruses/genetics , Simian Immunodeficiency Virus/genetics , Animals , Base Sequence , Cells, Cultured , DNA, Viral/blood , Genome, Viral , Humans , Lung/drug effects , Lung/virology , Lymph Nodes/drug effects , Lymph Nodes/virology , Macaca mulatta , Proviruses/drug effects , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/virology , Spleen/pathology , Spleen/virology , Tissue Distribution , Transgenes
19.
J Alzheimers Dis ; 78(3): 855-869, 2020.
Article in English | MEDLINE | ID: mdl-33074235

ABSTRACT

Across the fields of virology and neuroscience, the role of neurotropic viruses in Alzheimer's disease (AD) has received renewed enthusiasm, with a particular focus on human herpesviruses (HHVs). Recent genomic analyses of brain tissue collections and investigations of the antimicrobial responses of amyloid-ß do not exclude a role of HHVs in contributing to or accelerating AD pathogenesis. Due to continued expansion in our aging cohort and the lack of effective treatments for AD, this composition examines a potential neuroviral theory of AD in light of these recent data. Consideration reveals a possible viral "Hit-and-Run" scenario of AD, as well as neurobiological mechanisms (i.e., neuroinflammation, protein quality control, oxidative stress) that may increase risk for AD following neurotropic infection. Although limitations exist, this theoretical framework reveals several novel therapeutic targets that may prove efficacious in AD.


Subject(s)
Alzheimer Disease/virology , Brain/metabolism , Genome, Viral , Herpesviridae Infections/metabolism , Herpesviridae/genetics , Inflammation/metabolism , Oxidative Stress , Animals , Brain/virology , DNA, Viral , Herpesviridae Infections/virology , Host Microbial Interactions , Humans , In Vitro Techniques , Inflammation/virology , Latent Infection , Viral Tropism
20.
Neurochem Int ; 141: 104880, 2020 12.
Article in English | MEDLINE | ID: mdl-33065212

ABSTRACT

The dysregulation of lipid homeostasis is emerging as a hallmark of many CNS diseases. As aberrant protein regulation is suggested to be a shared pathological feature amongst many neurodegenerative conditions, such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), disruptions in neuronal lipid processing may contribute to disease progression in the CNS. Specifically, given the endoplasmic reticulum (ER) dual role in lipid homeostasis as well as protein quality control (PQC) via unfolded protein response (UPR), lipid dysregulation in the CNS may converge on ER functioning and constitute a crucial mechanism underlying aberrant protein aggregation. In the current review, we discuss the diverse roles of lipid species as essential components of the CNS. Moreover, given the importance of both lipid dysregulation and protein aggregation in pathology of CNS diseases, we attempt to assess the potential downstream cross-talk between lipid dysregulation and ER dependent PQC mechanisms, with special focus on HIV-associated neurodegenerative disorders (HAND).


Subject(s)
AIDS Dementia Complex/metabolism , AIDS Dementia Complex/physiopathology , Endoplasmic Reticulum Stress , HIV Infections/physiopathology , Lipid Metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Animals , HIV-1 , Humans , Unfolded Protein Response
SELECTION OF CITATIONS
SEARCH DETAIL