Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
bioRxiv ; 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37425704

ABSTRACT

Granulocyte colony stimulating factor (G-CSF) is commonly used as adjunct treatment to hasten recovery from neutropenia following chemotherapy and autologous transplantation of hematopoietic stem and progenitor cells (HSPCs) for malignant disorders. However, the utility of G-CSF administration after ex vivo gene therapy procedures targeting human HSPCs has not been thoroughly evaluated. Here, we provide evidence that post-transplant administration of G-CSF impedes engraftment of CRISPR-Cas9 gene edited human HSPCs in xenograft models. G-CSF acts by exacerbating the p53-mediated DNA damage response triggered by Cas9- mediated DNA double-stranded breaks. Transient p53 inhibition in culture attenuates the negative impact of G-CSF on gene edited HSPC function. In contrast, post-transplant administration of G-CSF does not impair the repopulating properties of unmanipulated human HSPCs or HSPCs genetically engineered by transduction with lentiviral vectors. The potential for post-transplant G-CSF administration to aggravate HSPC toxicity associated with CRISPR-Cas9 gene editing should be considered in the design of ex vivo autologous HSPC gene editing clinical trials.

2.
Hum Gene Ther ; 33(23-24): 1293-1304, 2022 12.
Article in English | MEDLINE | ID: mdl-36094106

ABSTRACT

Ex vivo gene therapy procedures targeting hematopoietic stem and progenitor cells (HSPCs) predominantly utilize lentivirus-based vectors for gene transfer. We provide the first pre-clinical evidence of the therapeutic utility of a foamy virus vector (FVV) for the genetic correction of human leukocyte adhesion deficiency type 1 (LAD-1), an inherited primary immunodeficiency resulting from mutation of the ß2 integrin common chain, CD18. CD34+ HSPCs isolated from a severely affected LAD-1 patient were transduced under a current good manufacturing practice-compatible protocol with FVV harboring a therapeutic CD18 transgene. LAD-1-associated cellular chemotactic defects were ameliorated in transgene-positive, myeloid-differentiated LAD-1 cells assayed in response to a strong neutrophil chemoattractant in vitro. Xenotransplantation of vector-transduced LAD-1 HSPCs in immunodeficient (NSG) mice resulted in long-term (∼5 months) human cell engraftment within murine bone marrow. Moreover, engrafted LAD-1 myeloid cells displayed in vivo levels of transgene marking previously reported to ameliorate the LAD-1 phenotype in a large animal model of the disease. Vector insertion site analysis revealed a favorable vector integration profile with no overt evidence of genotoxicity. These results coupled with the unique biological features of wild-type foamy virus support the development of FVVs for ex vivo gene therapy of LAD-1.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome , Spumavirus , Humans , Mice , Animals , Spumavirus/genetics , Genetic Vectors/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/therapy , Hematopoietic Stem Cells , CD18 Antigens/genetics , Antigens, CD34/genetics
3.
PDA J Pharm Sci Technol ; 76(6): 474-484, 2022.
Article in English | MEDLINE | ID: mdl-35296562

ABSTRACT

Many medical conditions require chronic treatment with subcutaneous injectable biologics often exceeding 1.0 mL. However, subcutaneous administration of volumes of 2.0 mL or greater using a standard needle and syringe or auto-injector proves challenging, and patients often must administer two separate injections to achieve their full dose or endure injection times in excess of 10 s if using a mechanical autoinjector. In addition, needle-based injections often cause patient anxiety and discomfort. In this article, we describe an approach to meet these needs with a needle-free medication delivery device capable of rapidly delivering up to 2.0 mL with minimal discomfort. A pilot study was conducted with this needle-free injection system to evaluate the delivery of a 2.0 mL volume in human subjects. The results demonstrated that injections of up to 2.0 mL were well tolerated and often preferred over two separate 1.0 mL injections using the needle-free injection system.


Subject(s)
Drug Delivery Systems , Syringes , Humans , Pilot Projects , Injections, Subcutaneous , Pharmaceutical Preparations
4.
Drug Deliv ; 28(1): 1915-1922, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34541982

ABSTRACT

Needle-free injection is a desirable goal for many reasons, including reducing pain, anxiety, and eliminating safety risks associated with needle-stick injuries. However, development of a safe, reliable needle-free device optimized for at-home use has been met with many challenges. Portal Instruments Inc. has been developing needle-free medication delivery using a well-designed hand-held device, PRIME, that is safe, intuitive to use, and utilizes advanced electronic control of a focused, high velocity, pressurized liquid injection stream. The PRECISE II human study demonstrated that the PRIME needle-free injection system was safe, well tolerated, and strongly preferred by participants for self-injections over a standard needle and syringe. In addition, the study was able to be completed early for superiority following the success of the pre-defined interim analysis.


Subject(s)
Injections, Subcutaneous/instrumentation , Patient Preference , Adult , Cross-Over Studies , Female , Humans , Injections, Subcutaneous/adverse effects , Male , Middle Aged , Needles , Prospective Studies
5.
Stem Cell Reports ; 16(9): 2336-2350, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34450041

ABSTRACT

Activation of NOTCH signaling in human hematopoietic stem/progenitor cells (HSPCs) by treatment with an engineered Delta-like ligand (DELTA1ext-IgG [DXI]) has enabled ex vivo expansion of short-term HSPCs, but the effect on long-term repopulating hematopoietic stem cells (LTR-HSCs) remains uncertain. Here, we demonstrate that ex vivo culture of human adult HSPCs with DXI under low oxygen tension limits ER stress in LTR-HSCs and lineage-committed progenitors compared with normoxic cultures. A distinct HSC gene signature was upregulated in cells cultured with DXI in hypoxia and, after 21 days of culture, the frequency of LTR-HSCs increased 4.9-fold relative to uncultured cells and 4.2-fold compared with the normoxia + DXI group. NOTCH and hypoxia pathways intersected to maintain undifferentiated phenotypes in cultured HSPCs. Our work underscores the importance of mitigating ER stress perturbations to preserve functional LTR-HSCs in extended cultures and offers a clinically feasible platform for the expansion of human HSPCs.


Subject(s)
Cell Hypoxia , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Receptors, Notch/metabolism , Antigens, CD34/metabolism , Biomarkers , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Computational Biology/methods , Humans , Molecular Sequence Annotation , Receptors, Notch/genetics , Signal Transduction , Transcriptome
6.
Cells ; 10(4)2021 03 26.
Article in English | MEDLINE | ID: mdl-33810313

ABSTRACT

Diamond Blackfan Anemia (DBA) is a congenital macrocytic anemia associated with ribosomal protein haploinsufficiency. Ribosomal dysfunction delays globin synthesis, resulting in excess toxic free heme in erythroid progenitors, early differentiation arrest, and pure red cell aplasia. In this study, DBA induced pluripotent stem cell (iPSC) lines were generated from blood mononuclear cells of DBA patients with inactivating mutations in RPS19 and subjected to hematopoietic differentiation to model disease phenotypes. In vitro differentiated hematopoietic cells were used to investigate whether eltrombopag, an FDA-approved mimetic of thrombopoietin with robust intracellular iron chelating properties, could rescue erythropoiesis in DBA by restricting the labile iron pool (LIP) derived from excessive free heme. DBA iPSCs exhibited RPS19 haploinsufficiency, reduction in the 40S/60S ribosomal subunit ratio and early erythroid differentiation arrest in the absence of eltrombopag, compared to control isogenic iPSCs established by CRISPR/Cas9-mediated correction of the RPS19 point mutation. Notably, differentiation of DBA iPSCs in the presence of eltrombopag markedly improved erythroid maturation. Consistent with a molecular mechanism based on intracellular iron chelation, we observed that deferasirox, a clinically licensed iron chelator able to permeate into cells, also enhanced erythropoiesis in our DBA iPSC model. In contrast, erythroid maturation did not improve substantially in DBA iPSC differentiation cultures supplemented with deferoxamine, a clinically available iron chelator that poorly accesses LIP within cellular compartments. These findings identify eltrombopag as a promising new therapeutic to improve anemia in DBA.


Subject(s)
Anemia, Diamond-Blackfan/drug therapy , Anemia, Diamond-Blackfan/pathology , Benzoates/therapeutic use , Cell Differentiation , Erythroid Cells/pathology , Hydrazines/therapeutic use , Induced Pluripotent Stem Cells/pathology , Models, Biological , Pyrazoles/therapeutic use , Anemia, Diamond-Blackfan/genetics , Animals , Base Sequence , Benzoates/pharmacology , Cell Differentiation/drug effects , Cell Line , Erythroid Cells/drug effects , Erythropoiesis , Humans , Hydrazines/pharmacology , Induced Pluripotent Stem Cells/drug effects , Intracellular Space/metabolism , Iron/metabolism , Mice, Inbred NOD , Mice, SCID , Mutation/genetics , Pyrazoles/pharmacology
7.
Infect Dis Rep ; 13(1): 58-71, 2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440699

ABSTRACT

Contaminated surfaces and indoor environments are important sources of infectious spread within hospital and non-hospital facilities. Bacterial infections such as infections with Clostridioides (formerly Clostridium) difficile (C. difficile) and Staphylococcus aureus (S. aureus) and its antibiotic resistant strains continue to pose a significant risk to healthcare workers and patients. Additionally, the recent emergence of the coronavirus disease 2019 (COVID-19) pandemic, which is caused by the novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlights the need for safe and effective methods to decontaminate surfaces to control infection spread in hospitals and the community. To address these critical needs, we tested a photocatalytic reactor decontamination method to disinfect contaminated surfaces in a hospital and a laboratory setting. By placing the reactor in a test hospital room, growth of S. aureus and C. difficile were significantly reduced compared with a control room. Additionally, using a model enveloped positive-sense single-stranded RNA virus, dengue virus type 2 (DENV2), we showed that the use of the photocatalytic reactor reduces viral infectivity. Collectively, the results demonstrate the potential utility of photocatalytic reactors in reducing the spread of highly contagious bacterial and viral infections through contaminated surfaces and environments.

8.
Mol Ther ; 29(4): 1611-1624, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33309880

ABSTRACT

Ex vivo gene correction of hematopoietic stem and progenitor cells (HSPCs) has emerged as a promising therapeutic approach for treatment of inherited human blood disorders. Use of engineered nucleases to target therapeutic transgenes to their endogenous genetic loci addresses many of the limitations associated with viral vector-based gene replacement strategies, such as insertional mutagenesis, variable gene dosage, and ectopic expression. Common methods of nuclease-mediated site-specific integration utilize the homology-directed repair (HDR) pathway. However, these approaches are inefficient in HSPCs, where non-homologous end joining (NHEJ) is the primary DNA repair mechanism. Recently, a novel NHEJ-based approach to CRISPR-Cas9-mediated transgene knockin, known as homology-independent targeted integration (HITI), has demonstrated improved site-specific integration frequencies in non-dividing cells. Here we utilize a HITI-based approach to achieve robust site-specific transgene integration in human mobilized peripheral blood CD34+ HSPCs. As proof of concept, a reporter gene was targeted to a clinically relevant genetic locus using a recombinant adeno-associated virus serotype 6 vector and single guide RNA/Cas9 ribonucleoprotein complexes. We demonstrate high levels of stable HITI-mediated genome editing (∼21%) in repopulating HSPCs after transplantation into immunodeficient mice. Our study demonstrates that HITI-mediated genome editing provides an effective alternative to HDR-based transgene integration in CD34+ HSPCs.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Therapy , Hematologic Diseases/genetics , Hematopoietic Stem Cell Transplantation , Animals , DNA End-Joining Repair/genetics , DNA Repair/genetics , Dependovirus/genetics , Gene Editing , Genetic Vectors/genetics , Genome, Human/genetics , Hematologic Diseases/pathology , Hematologic Diseases/therapy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , RNA, Guide, Kinetoplastida/genetics , Recombinational DNA Repair/genetics , Stem Cells/cytology , Stem Cells/metabolism
9.
Genes (Basel) ; 11(12)2020 12 13.
Article in English | MEDLINE | ID: mdl-33322084

ABSTRACT

CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9)-mediated genome editing holds remarkable promise for the treatment of human genetic diseases. However, the possibility of off-target Cas9 activity remains a concern. To address this issue using clinically relevant target cells, we electroporated Cas9 ribonucleoprotein (RNP) complexes (independently targeted to two different genomic loci, the CXCR4 locus on chromosome 2 and the AAVS1 locus on chromosome 19) into human mobilized peripheral blood-derived hematopoietic stem and progenitor cells (HSPCs) and assessed the acquisition of somatic mutations in an unbiased, genome-wide manner via whole genome sequencing (WGS) of single-cell-derived HSPC clones. Bioinformatic analysis identified >20,000 total somatic variants (indels, single nucleotide variants, and structural variants) distributed among Cas9-treated and non-Cas9-treated control HSPC clones. Statistical analysis revealed no significant difference in the number of novel non-targeted indels among the samples. Moreover, data analysis showed no evidence of Cas9-mediated indel formation at 623 predicted off-target sites. The median number of novel single nucleotide variants was slightly elevated in Cas9 RNP-recipient sample groups compared to baseline, but did not reach statistical significance. Structural variants were rare and demonstrated no clear causal connection to Cas9-mediated gene editing procedures. We find that the collective somatic mutational burden observed within Cas9 RNP-edited human HSPC clones is indistinguishable from naturally occurring levels of background genetic heterogeneity.


Subject(s)
CRISPR-Cas Systems , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 2/genetics , Clone Cells , Gene Editing , Hematopoietic Stem Cells , Adult , Female , Genetic Loci , Humans , Receptors, CXCR4/genetics
11.
Blood Adv ; 4(2): 367-379, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31985806

ABSTRACT

Acute myeloid leukemia (AML) is a genetically heterogeneous disease that is characterized by abnormal clonal proliferation of myeloid progenitor cells found predominantly within the bone marrow (BM) and blood. Recent studies suggest that genetic and phenotypic alterations in the BM microenvironment support leukemogenesis and allow leukemic cells to survive and evade chemotherapy-induced death. However, despite substantial evidence indicating the role of tumor-host interactions in AML pathogenesis, little is known about the complex microenvironment of the BM. To address this, we performed novel proteomic profiling of the noncellular compartment of the BM microenvironment in patients with AML (n = 10) and age- and sex-matched healthy control subjects (n = 10) using an aptamer-based, highly multiplexed, affinity proteomics platform (SOMAscan). We show that proteomic assessment of blood or RNA-sequencing of BM are suboptimal alternate screening strategies to determine the true proteomic composition of the extracellular soluble compartment of AML patient BM. Proteomic analysis revealed that 168 proteins significantly differed in abundance, with 91 upregulated and 77 downregulated in leukemic BM. A highly connected signaling network of cytokines and chemokines, including IL-8, was found to be the most prominent proteomic signature associated with AML in the BM microenvironment. We report the first description of significantly elevated levels of the myelosuppressive chemokine CCL23 (myeloid progenitor inhibitory factor-1) in both AML and myelodysplastic syndrome patients and perform functional experiments supportive of a role in the suppression of normal hematopoiesis. This unique paired RNA-sequencing and proteomics data set provides innovative mechanistic insights into AML and healthy aging and should serve as a useful public resource.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Proteomics/methods , Case-Control Studies , Cellular Microenvironment , Chemokines/analysis , Chemokines, CC/metabolism , Cytokines/analysis , Gene Expression Regulation, Leukemic , Humans , Interleukin-8/metabolism , Neoplasm Proteins/analysis
12.
Exp Hematol ; 73: 1-6.e6, 2019 05.
Article in English | MEDLINE | ID: mdl-30986494

ABSTRACT

A causal link between hematopoietic stem/progenitor cell (HSPC) dysfunction and DNA damage accrual has been proposed. Clinically relevant strategies to maintain genome integrity in these cells are needed. Here we report that eltrombopag, a small molecule agonist of the thrombopoietin (TPO) receptor used in the clinic, promotes DNA double-strand break (DSB) repair in human HSPCs. We found that eltrombopag specifically activates the classic nonhomologous end-joining (C-NHEJ) DNA repair mechanism, a pathway known to support genome integrity. Eltrombopag-mediated DNA repair results in enhanced genome stability, survival, and function of primary human HSPCs, as demonstrated in karyotyping analyses, colony-forming unit assays and after transplantation in immunodeficient NSG mice. Eltrombopag may offer a new therapeutic modality to protect human HSPCs against genome insults.


Subject(s)
Benzoates/pharmacology , DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , Hematopoietic Stem Cells/metabolism , Hydrazines/pharmacology , Pyrazoles/pharmacology , Cells, Cultured , Humans , Receptors, Thrombopoietin/metabolism
13.
J Neurointerv Surg ; 11(2): 127-132, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29930159

ABSTRACT

BACKGROUND: Stent retriever thrombectomy (SRT) in acute thromboembolic stroke can result in post-thrombectomy subarachnoid hemorrhage (PTSAH). Intraprocedural findings associated with PTSAH are not well defined. OBJECTIVE: To identify angiographic findings and procedural factors during SRT that are associated with PTSAH. MATERIALS AND METHODS: This was a retrospective, observational cohort study of consecutive patients with middle cerebral artery (MCA) acute ischemic stroke treated with SRT. Inclusion criteria were: (1) age ≥18 years; (2) thromboembolic occlusion of the MCA; (3) at least one stent retriever pass beginning in an M2 branch; (4) postprocedural CT or MRI scan within 24 hours; (5) non-enhanced CT Alberta Stroke Program Early CT Score >5. Exclusion criteria included multi-territory stroke before SRT. RESULTS: Eighty-five patients were enrolled; eight patients had PTSAH (group 1) and 77 did not (group 2). Baseline demographic and clinical characteristics were comparable between the two groups. In group 1, a significantly greater proportion of patients had more than two stent retriever passes (62.5% vs 18.2%, P=0.01), a stent retriever positioned ≥2 cm along an M2 branch (100% vs 30.2%, P=0.002), and the presence of severe iatrogenic vasospasm before SRT pass (37.5% vs 5.2%, P=0.02). One patient with PTSAH and associated mass effect deteriorated clinically. CONCLUSIONS: An increased number of stent retriever passes, distal device positioning, and presence of severe vasospasm were associated with PTSAH. Neurological deterioration with PTSAH can occur.


Subject(s)
Infarction, Middle Cerebral Artery/diagnostic imaging , Intraoperative Neurophysiological Monitoring/methods , Stents , Stroke/diagnostic imaging , Subarachnoid Hemorrhage/diagnostic imaging , Thrombectomy/adverse effects , Adolescent , Adult , Aged , Cohort Studies , Female , Humans , Infarction, Middle Cerebral Artery/surgery , Intraoperative Neurophysiological Monitoring/trends , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Retrospective Studies , Stroke/surgery , Subarachnoid Hemorrhage/etiology , Thrombectomy/trends , Young Adult
14.
Front Surg ; 6: 73, 2019.
Article in English | MEDLINE | ID: mdl-31998742

ABSTRACT

Complete spinal cord injury is a devastating occurrence afflicting millions of people worldwide with no available treatment for functional motor recovery. In this report, we describe a procedure in which we used parts of a device available for chronic pain treatment to provide a neuromodulation of motor nerve roots in a case with complete motor and sensory paraplegia. By using a retrograde trans-foraminal approach to implant electrodes close to L2-S1 motor nerve roots bilaterally, we were able to stimulate those nerves and induce precise movements at the joints of lower extremity in a T5 complete spinal cord injury case. We believe that our approach shows potential of the device as a rehabilitation system with the possibility of a parallel electric circuitry that can bridge a damaged spinal cord.

15.
Mol Ther Methods Clin Dev ; 3: 16004, 2016.
Article in English | MEDLINE | ID: mdl-27722179

ABSTRACT

Compared to other integrating viral vectors, foamy virus (FV) vectors have distinct advantages as a gene transfer tool, including their nonpathogenicity, the ability to carry larger transgene cassettes, and increased stability of virus particles due to DNA genome formation within the virions. Proof of principle of its therapeutic utility was provided with the correction of canine leukocyte adhesion deficiency using autologous CD34+ cells transduced with FV vector carrying the canine CD18 gene, demonstrating its long-term safety and efficacy. However, infectious titers of FV-human(h)CD18 were low and not suitable for manufacturing of clinical-grade product. Herein, we developed a scalable production and purification process that resulted in 60-fold higher FV-hCD18 titers from ~1.7 × 104 to 1.0 × 106 infectious units (IU)/ml. Process development improvements included use of polyethylenimine-based transfection, use of a codon-optimized gag, heparin affinity chromatography, tangential flow filtration, and ultracentrifugation, which reproducibly resulted in 5,000-fold concentrated and purified virus, an overall yield of 19 ± 3%, and final titers of 1-2 × 109 IU/ml. Highly concentrated vector allowed reduction of final dimethyl sulfoxide (DMSO) concentration, thereby avoiding DMSO-induced toxicity to CD34+ cells while maintaining high transduction efficiencies. This process development results in clinically relevant, high titer FV which can be scaled up for clinical grade production.

16.
Sci Rep ; 6: 28965, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27377618

ABSTRACT

Germline endogenous viral elements (EVEs) genetically preserve viral nucleotide sequences useful to the study of viral evolution, gene mutation, and the phylogenetic relationships among host organisms. Here, we describe a lineage-specific, adeno-associated virus (AAV)-derived endogenous viral element (mAAV-EVE1) found within the germline of numerous closely related marsupial species. Molecular screening of a marsupial DNA panel indicated that mAAV-EVE1 occurs specifically within the marsupial suborder Macropodiformes (present-day kangaroos, wallabies, and related macropodoids), to the exclusion of other Diprotodontian lineages. Orthologous mAAV-EVE1 locus sequences from sixteen macropodoid species, representing a speciation history spanning an estimated 30 million years, facilitated compilation of an inferred ancestral sequence that recapitulates the genome of an ancient marsupial AAV that circulated among Australian metatherian fauna sometime during the late Eocene to early Oligocene. In silico gene reconstruction and molecular modelling indicate remarkable conservation of viral structure over a geologic timescale. Characterisation of AAV-EVE loci among disparate species affords insight into AAV evolution and, in the case of macropodoid species, may offer an additional genetic basis for assignment of phylogenetic relationships among the Macropodoidea. From an applied perspective, the identified AAV "fossils" provide novel capsid sequences for use in translational research and clinical applications.


Subject(s)
Dependovirus/classification , Dependovirus/genetics , Fossils , Germ Cells/virology , Marsupialia/virology , Animals , Computational Biology , Evolution, Molecular
17.
Cogn Emot ; 29(6): 1007-25, 2015.
Article in English | MEDLINE | ID: mdl-25297882

ABSTRACT

Previous research has yielded inconsistent findings concerning the relationship between envy and schadenfreude. Three studies examined whether the distinction between benign and malicious envy can resolve this inconsistency. We found that malicious envy is related to schadenfreude, while benign envy is not. This result held both in the Netherlands where benign and malicious envy are indicated by separate words (Study 1: Sample A, N = 139; Sample B, N = 150), and in the USA where a single word is used to denote both types (Study 2, N = 180; Study 3, N = 349). Moreover, the effect of malicious envy on schadenfreude was independent of other antecedents of schadenfreude (such as feelings of inferiority, disliking the target person, anger, and perceived deservedness). These findings improve our understanding of the antecedents of schadenfreude and help reconcile seemingly contradictory findings on the relationship between envy and schadenfreude.


Subject(s)
Emotions , Social Behavior , Adolescent , Adult , Female , Humans , Male , Middle Aged , Netherlands , United States , Young Adult
18.
Mol Ther ; 22(11): 1923-35, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25200009

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disorder caused by mutations in the dystrophin gene, without curative treatment yet available. Our study provides, for the first time, the overall safety profile and therapeutic dose of a recombinant adeno-associated virus vector, serotype 8 (rAAV8) carrying a modified U7snRNA sequence promoting exon skipping to restore a functional in-frame dystrophin transcript, and injected by locoregional transvenous perfusion of the forelimb. Eighteen Golden Retriever Muscular Dystrophy (GRMD) dogs were exposed to increasing doses of GMP-manufactured vector. Treatment was well tolerated in all, and no acute nor delayed adverse effect, including systemic and immune toxicity was detected. There was a dose relationship for the amount of exon skipping with up to 80% of myofibers expressing dystrophin at the highest dose. Similarly, histological, nuclear magnetic resonance pathological indices and strength improvement responded in a dose-dependent manner. The systematic comparison of effects using different independent methods, allowed to define a minimum threshold of dystrophin expressing fibers (>33% for structural measures and >40% for strength) under which there was no clear-cut therapeutic effect. Altogether, these results support the concept of a phase 1/2 trial of locoregional delivery into upper limbs of nonambulatory DMD patients.


Subject(s)
Dependovirus/genetics , Dystrophin/genetics , Forelimb/physiopathology , Muscular Dystrophy, Duchenne/therapy , RNA, Small Nuclear/genetics , Animals , Cohort Studies , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Exons , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Infusions, Intravenous , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , RNA, Small Nuclear/metabolism
19.
Soc Neurosci ; 8(6): 640-9, 2013.
Article in English | MEDLINE | ID: mdl-24138447

ABSTRACT

The misfortunes of enviable individuals are met by observers with pleasure whereas those of "average", non-enviable individuals elicit pain. These responses are mirrored in deservingness judgments, as enviable individuals' misfortunes are perceived as deserved and those of non-enviable individuals perceived as undeserved. However, the neural underpinnings of these deservingness disparities remain unknown. To explore this phenomenon, we utilized fMRI to test the hypotheses that (A) non-enviable targets' misfortunes would be associated with activation of brain regions that mediate empathic responding (pain matrix, mentalizing network) and not for enviable targets and (B) that activation of those regions would predict decreases in deservingness judgments. Supporting our first hypothesis, the misfortunes of non-enviable targets (as opposed to good fortunes) were associated with activation of the mentalizing network: medial prefrontal cortex, posterior cingulate cortex, temporal-parietal junction, and anterior temporal lobes. Supporting our second hypothesis, dorsomedial prefrontal cortex activation from this contrast was negatively correlated with subsequent reports of how much the non-enviable target deserved his/her misfortune. These findings suggest that non-enviable individuals' misfortunes are perceived as unjust due, in part, to the recruitment of the mentalizing network.


Subject(s)
Brain Mapping , Brain/physiology , Jealousy , Social Justice , Theory of Mind/physiology , Adolescent , Empathy/physiology , Female , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging , Male , Social Justice/psychology , Young Adult
20.
PLoS One ; 8(8): e69879, 2013.
Article in English | MEDLINE | ID: mdl-23936358

ABSTRACT

Conventional non-viral gene transfer uses bacterial plasmid DNA containing antibiotic resistance genes, cis-acting bacterial sequence elements, and prokaryotic methylation patterns that may adversely affect transgene expression and vector stability in vivo. Here, we describe novel replicative forms of a eukaryotic vector DNA that consist solely of an expression cassette flanked by adeno-associated virus (AAV) inverted terminal repeats. Extensive structural analyses revealed that this AAV-derived vector DNA consists of linear, duplex molecules with covalently closed ends (termed closed-ended, linear duplex, or "CELiD", DNA). CELiD vectors, produced in Sf9 insect cells, require AAV rep gene expression for amplification. Amounts of CELiD DNA produced from insect cell lines stably transfected with an ITR-flanked transgene exceeded 60 mg per 5 × 10(9) Sf9 cells, and 1-15 mg from a comparable number of parental Sf9 cells in which the transgene was introduced via recombinant baculovirus infection. In mice, systemically delivered CELiD DNA resulted in long-term, stable transgene expression in the liver. CELiD vectors represent a novel eukaryotic alternative to bacterial plasmid DNA.


Subject(s)
DNA, Recombinant/genetics , DNA/genetics , Dependovirus/genetics , Dependovirus/physiology , Genome, Viral/genetics , Transfection/methods , Virus Replication , Animals , Genetic Engineering , Genetic Vectors/genetics , Male , Mice , Sf9 Cells , Spodoptera , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...