Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Article in English | MEDLINE | ID: mdl-36690462

ABSTRACT

X-linked retinoschisis (XLRS) is an inherited vitreoretinal dystrophy causing visual impairment in males starting at a young age with an estimated prevalence of 1:5000 to 1:25,000. The condition was first observed in two affected brothers by Josef Haas in 1898 and is clinically diagnosed by characteristic intraretinal cysts arranged in a petaloid "spoke-wheel" pattern centered in the macula. When clinical electroretinogram (ERG) testing began in the 1960s, XLRS was noted to have a characteristic reduction of the dark-adapted b-wave amplitude despite normal or usually nearly normal a-wave amplitudes, which became known as the "electronegative ERG response" of XLRS disease. The causative gene, RS1, was identified on the X-chromosome in 1997 and led to understanding the molecular and cellular basis of the condition, discerning the structure and function of the retinoschisin protein, and generating XLRS murine models. Along with parallel development of gene delivery vectors suitable for targeting retinal diseases, successful gene augmentation therapy was demonstrated by rescuing the XLRS phenotype in mouse. Two human phase I/II therapeutic XLRS gene augmentation studies were initiated; and although these did not yield definitive improvement in visual function, they gave significant new knowledge and experience, which positions the field for further near-term clinical testing with enhanced, next-generation gene therapy for XLRS patients.


Subject(s)
Retinoschisis , Male , Humans , Animals , Mice , Retinoschisis/genetics , Retinoschisis/therapy , Retinoschisis/diagnosis , Mutation , Electroretinography , Phenotype , Genetic Therapy , Eye Proteins/genetics , Eye Proteins/metabolism , Retina/metabolism
2.
Nat Commun ; 13(1): 5487, 2022 09 19.
Article in English | MEDLINE | ID: mdl-36123353

ABSTRACT

Relapsed or refractory pediatric acute myeloid leukemia (AML) is associated with poor outcomes and relapse risk prediction approaches have not changed significantly in decades. To build a robust transcriptional risk prediction model for pediatric AML, we perform RNA-sequencing on 1503 primary diagnostic samples. While a 17 gene leukemia stem cell signature (LSC17) is predictive in our aggregated pediatric study population, LSC17 is no longer predictive within established cytogenetic and molecular (cytomolecular) risk groups. Therefore, we identify distinct LSC signatures on the basis of AML cytomolecular subtypes (LSC47) that were more predictive than LSC17. Based on these findings, we build a robust relapse prediction model within a training cohort and then validate it within independent cohorts. Here, we show that LSC47 increases the predictive power of conventional risk stratification and that applying biomarkers in a manner that is informed by cytomolecular profiling outperforms a uniform biomarker approach.


Subject(s)
Gene Expression Profiling , Leukemia, Myeloid, Acute , Biomarkers , Child , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells , RNA , Recurrence
4.
Front Genet ; 12: 665888, 2021.
Article in English | MEDLINE | ID: mdl-34149808

ABSTRACT

RNA sequencing (RNAseq) has been widely used to generate bulk gene expression measurements collected from pools of cells. Only relatively recently have single-cell RNAseq (scRNAseq) methods provided opportunities for gene expression analyses at the single-cell level, allowing researchers to study heterogeneous mixtures of cells at unprecedented resolution. Tumors tend to be composed of heterogeneous cellular mixtures and are frequently the subjects of such analyses. Extensive method developments have led to several protocols for scRNAseq but, owing to the small amounts of RNA in single cells, technical constraints have required compromises. For example, the majority of scRNAseq methods are limited to sequencing only the 3' or 5' termini of transcripts. Other protocols that facilitate full-length transcript profiling tend to capture only polyadenylated mRNAs and are generally limited to processing only 96 cells at a time. Here, we address these limitations and present a novel protocol that allows for the high-throughput sequencing of full-length, total RNA at single-cell resolution. We demonstrate that our method produced strand-specific sequencing data for both polyadenylated and non-polyadenylated transcripts, enabled the profiling of transcript regions beyond only transcript termini, and yielded data rich enough to allow identification of cell types from heterogeneous biological samples.

5.
Mol Ther ; 29(6): 2030-2040, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33601057

ABSTRACT

This study explored systemic immune changes in 11 subjects with X-linked retinoschisis (XLRS) in a phase I/IIa adeno-associated virus 8 (AAV8)-RS1 gene therapy trial (ClinicalTrials.gov: NCT02317887). Immune cell proportions and serum analytes were compared to 12 healthy male controls. At pre-dosing baseline the mean CD4/CD8 ratio of XLRS subjects was elevated. CD11c+ myeloid dendritic cells (DCs) and the serum epidermal growth factor (EGF) level were decreased, while CD123+ plasmacytoid DCs and serum interferon (IFN)-γ and tumor necrosis factor (TNF)-α were increased, indicating that the XLRS baseline immune status differs from that of controls. XLRS samples 14 days after AAV8-RS1 administration were compared with the XLRS baseline. Frequency of CD11b+CD11c+ DCc was decreased in 8 of 11 XLRS subjects across all vector doses (1e9-3e11 vector genomes [vg]/eye). CD8+human leukocyte antigen-DR isotype (HLA-DR)+ cytotoxic T cells and CD68+CD80+ macrophages were upregulated in 10 of 11 XLRS subjects, along with increased serum granzyme B in 8 of 11 XLRS subjects and elevated IFN-γ in 9 of 11 XLRS subjects. The six XLRS subjects with ocular inflammation after vector application gave a modestly positive correlation of inflammation score to their respective baseline CD4/CD8 ratios. This exploratory study indicates that XLRS subjects may exhibit a proinflammatory, baseline immune phenotype, and that intravitreal dosing with AAV8-RS1 leads to systemic immune activation with an increase of activated lymphocytes, macrophages, and proinflammatory cytokines.


Subject(s)
Eye Proteins/genetics , Genetic Diseases, X-Linked/etiology , Genetic Diseases, X-Linked/therapy , Genetic Therapy , Retinoschisis/genetics , Retinoschisis/immunology , Retinoschisis/therapy , Cytokines/blood , Cytokines/metabolism , Dependovirus/genetics , Disease Management , Genetic Predisposition to Disease , Genetic Therapy/methods , Genetic Vectors , Humans , Immunity , Immunity, Cellular , Retinoschisis/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome
6.
Transl Vis Sci Technol ; 9(7): 28, 2020 06.
Article in English | MEDLINE | ID: mdl-32844051

ABSTRACT

Purpose: Electric micro-current has been shown to enhance penetration and transduction of adeno-associated viral (AAV) vectors in mouse retina after intravitreal administration. We termed this: "electric-current vector mobility (ECVM)." The present study considered whether ECVM could augment retinal transduction efficiency of intravitreal AAV8-CMV-EGFP in normal rabbit and nonhuman primate (NHP) macaque. Potential mechanisms underlying enhanced retinal transduction by ECVM were also studied. Methods: We applied an electric micro-current across the intact eye of normal rabbit and monkey in vivo for a brief period immediately after intravitreal injection of AAV8-CMV-EGFP. Retinal GFP expression was evaluated by fundus imaging in vivo. Retinal immunohistochemistry was performed to assess the distribution of retinal cells transduced by the AAV8-EGFP. Basic fibroblast growth factor (bFGF) was analyzed by quantitative RT-polymerase chain reaction (PCR). Müller glial reactivity and inner limiting membrane (ILM) were examined by the glial fibrillary acidic protein (GFAP) and vimentin staining in mouse retina, respectively. Results: ECVM significantly increased the efficiency of AAV reaching and transducing the rabbit retina following intravitreal injection, with gene expression in inner nuclear layer, ganglion cells, and Müller cells. Similar trend of improvement was observed in the ECVM-treated monkey eye. The electric micro-current upregulated bFGF expression in Müller cells and vimentin showed ILM structural changes in mouse retina. Conclusions: ECVM promotes the transduction efficiency of AAV8-CMV-GFP in normal rabbit and monkey retinas following intravitreal injection. Translational Relevance: This work has potential translational relevance to human ocular gene therapy by increasing retinal expression of therapeutic vectors given by intravitreal administration.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Dependovirus/genetics , Gene Expression , Genetic Vectors/genetics , Rabbits , Retina , Transduction, Genetic
7.
Mol Ther Methods Clin Dev ; 18: 422-427, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32695844

ABSTRACT

Intravitreal administration for human adeno-associated vector (AAV) delivery is easier and less traumatic to ocular tissues than subretinal injection, but it gives limited retinal transduction. AAV vectors are large (about 4,000 kDa) compared with most intraocular drugs, such as ranibizumab (48 kDa), and the large size impedes diffusion to reach the retina from the usual injection site in the anterior/mid-vitreous. Intuitively, a preferred placement for the vector would be deep in the vitreous near the retina, which we term "para-retinal" delivery. We explored the consequences of para-retinal intravitreal delivery in the rabbit eye and in non-human primate (NHP) eye. 1 h after para-retinal administration in the rabbit eye, the vector concentration near the retina remained four times greater than in the anterior vitreous, indicating limited vector diffusion through the gelatinous vitreous matrix. In NHP, para-retinal placement showed greater transduction in the fovea than vector applied in the mid-vitreous. More efficient retinal delivery translates to using lower vector doses, with reduced risk of ocular inflammatory exposure. These results indicate that para-retinal delivery yields more effective vector concentration near the retina, thereby increasing the potential for better retinal transduction in human clinical application.

8.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31816636

ABSTRACT

Lung group 2 innate lymphoid cells (ILC2s) drive allergic inflammation and promote tissue repair. ILC2 development is dependent on the transcription factor retinoic acid receptor-related orphan receptor (RORα), which is also expressed in common ILC progenitors. To elucidate the developmental pathways of lung ILC2s, we generated RORα lineage tracer mice and performed single-cell RNA sequencing, flow cytometry, and functional analyses. In adult mouse lungs, we found an IL-18Rα+ST2- population different from conventional IL-18Rα-ST2+ ILC2s. The former was GATA-3intTcf7EGFP+Kit+, produced few cytokines, and differentiated into multiple ILC lineages in vivo and in vitro. In neonatal mouse lungs, three ILC populations were identified, namely an ILC progenitor population similar to that in adult lungs and two distinct effector ILC2 subsets that differentially produced type 2 cytokines and amphiregulin. Lung ILC progenitors might actively contribute to ILC-poiesis in neonatal and inflamed adult lungs. In addition, neonatal lung ILC2s include distinct proinflammatory and tissue-repairing subsets.


Subject(s)
Immunity, Innate/immunology , Lung/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , Stem Cells/immunology , Amphiregulin/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cytokines/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Single-Cell Analysis/methods
9.
Cell Rep ; 29(8): 2338-2354.e7, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31708418

ABSTRACT

Extra-cranial malignant rhabdoid tumors (MRTs) and cranial atypical teratoid RTs (ATRTs) are heterogeneous pediatric cancers driven primarily by SMARCB1 loss. To understand the genome-wide molecular relationships between MRTs and ATRTs, we analyze multi-omics data from 140 MRTs and 161 ATRTs. We detect similarities between the MYC subgroup of ATRTs (ATRT-MYC) and extra-cranial MRTs, including global DNA hypomethylation and overexpression of HOX genes and genes involved in mesenchymal development, distinguishing them from other ATRT subgroups that express neural-like features. We identify five DNA methylation subgroups associated with anatomical sites and SMARCB1 mutation patterns. Groups 1, 3, and 4 exhibit cytotoxic T cell infiltration and expression of immune checkpoint regulators, consistent with a potential role for immunotherapy in rhabdoid tumor patients.


Subject(s)
Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Child , DNA Methylation/genetics , DNA Methylation/physiology , Female , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mutation/genetics , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Skull Base Neoplasms/metabolism , Skull Base Neoplasms/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Teratoma/metabolism , Teratoma/pathology
10.
Mol Ther ; 26(9): 2282-2294, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30196853

ABSTRACT

This study evaluated the safety and tolerability of ocular RS1 adeno-associated virus (AAV8-RS1) gene augmentation therapy to the retina of participants with X-linked retinoschisis (XLRS). XLRS is a monogenic trait affecting only males, caused by mutations in the RS1 gene. Retinoschisin protein is secreted principally in the outer retina, and its absence results in retinal cavities, synaptic dysfunction, reduced visual acuity, and susceptibility to retinal detachment. This phase I/IIa single-center, prospective, open-label, three-dose-escalation clinical trial administered vector to nine participants with pathogenic RS1 mutations. The eye of each participant with worse acuity (≤63 letters; Snellen 20/63) received the AAV8-RS1 gene vector by intravitreal injection. Three participants were assigned to each of three dosage groups: 1e9 vector genomes (vg)/eye, 1e10 vg/eye, and 1e11 vg/eye. The investigational product was generally well tolerated in all but one individual. Ocular events included dose-related inflammation that resolved with topical and oral corticosteroids. Systemic antibodies against AAV8 increased in a dose-related fashion, but no antibodies against RS1 were observed. Retinal cavities closed transiently in one participant. Additional doses and immunosuppressive regimens are being explored to pursue evidence of safety and efficacy (ClinicalTrials.gov: NCT02317887).


Subject(s)
Eye Proteins/metabolism , Genetic Therapy/methods , Retinoschisis/therapy , Adult , Aged , Eye Proteins/genetics , Female , Humans , Intravitreal Injections , Male , Middle Aged , Mutation/genetics , Retina/metabolism , Retina/pathology , Retinoschisis/genetics , Retinoschisis/metabolism , Young Adult
11.
Mol Ther Methods Clin Dev ; 5: 16011, 2016.
Article in English | MEDLINE | ID: mdl-27626041

ABSTRACT

X-linked retinoschisis (XLRS) is a retinal disease caused by mutations in the gene encoding the protein retinoschisin (RS1) and is one of the most common causes of macular degeneration in young men. Our therapeutic approach for XLRS is based on the administration of AAV8-scRS/IRBPhRS, an adeno-associated viral vector coding the human RS1 protein, via the intravitreal (IVT) route. Two Good Laboratory Practice studies, a 9-month study in New Zealand White rabbits (n = 124) injected with AAV8-scRS/IRBPhRS at doses of 2E9, 2E10, 2E11, and 1.5E12 vector genomes/eye (vg/eye), and a 6-month study in Rs1-KO mice (n = 162) dosed with 2E9 and 2E10 vg/eye of the same vector were conducted to assess ocular and systemic safety. A self-resolving, dose-dependent vitreal inflammation was the main ocular finding, and except for a single rabbit dosed with 1.5E12 vg/eye, which showed a retinal detachment, no other ocular adverse event was reported. Systemic toxicity was not identified in either species. Biodistribution analysis in Rs1-KO mice detected spread of vector genome in extraocular tissues, but no evidence of organ or tissues damage was found. These studies indicate that IVT administration of AAV8-scRS/IRBPhRS is safe and well tolerated and support its advancement into a phase 1/2a clinical trial for XLRS.

12.
Exp Hematol ; 44(7): 635-40, 2016 07.
Article in English | MEDLINE | ID: mdl-27090409

ABSTRACT

The critical human cells that produce neutrophils and platelets within 3 weeks in recipients of hematopoietic transplants are thought to produce these mature blood cells with the same kinetics in sublethally irradiated immunodeficient mice. Quantification of their numbers indicates their relative underrepresentation in cord blood (CB), likely explaining the clinical inadequacy of single CB units in rescuing hematopoiesis in myelosuppressed adult patients. We here describe that exposure of CD34(+) CB cells ex vivo to growth factors that markedly expand their numbers and colony-forming cell content also rapidly (within 24 hours) produce a significant and sustained net loss of their original short-term repopulating activity. This loss of short-term in vivo repopulating activity affects early platelet production faster than early neutrophil output, consistent with their origin from distinct input populations. Moreover, this growth factor-mediated loss is not abrogated by published strategies to increase progenitor homing despite evidence that the effect on rapid neutrophil production is paralleled in time and amount by a loss of the homing of their committed clonogenic precursors to the bone marrow. These results highlight the inability of in vitro or phenotype assessments to reliably predict clinical engraftment kinetics of cultured CB cells.


Subject(s)
Blood Platelets/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myelopoiesis , Neutrophils/metabolism , Thrombopoiesis , Animals , Cell Differentiation/drug effects , Cells, Cultured , Graft Survival , Hematopoietic Cell Growth Factors/metabolism , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Myelopoiesis/drug effects , Thrombopoiesis/drug effects
13.
Invest Ophthalmol Vis Sci ; 56(11): 6810-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26567794

ABSTRACT

PURPOSE: Ciliary neurotrophic factor (CNTF) was recently shown to augment cone function in CNGB3 mutant achromat dogs. However, testing CNTF-releasing implant in human CNGB3 achromats failed to show benefit. We evaluated the effects of CNTF protein on the retinal function in an additional achromatopsia model, the CNGB3-/- mouse. METHODS: Fifty-nine CNGB3-/- mice (postnatal day [PD] ± SD = 30 ± 7) received a unilateral intravitreal injection of 1 or 2 µg CNTF protein, and 15 wild-type (WT) mice (PD = 34 ± 3) received 1 µg CNTF. Retinal function was evaluated by flash ERG and photopic flicker ERG (fERG) at 7 and 14 days after treatment. RESULTS: Seven days post CNTF, the photopic b-wave Vmax was significantly increased in CNGB3-/- mice (P < 0.01), whereas it was reduced in WT mice (P < 0.05). Ciliary neurotrophic factor significantly increased the amplitude of photopic fERG and the photopic oscillatory potentials (OPs) in CNGB3-/- mice. Ciliary neurotrophic factor did not alter the scotopic a-wave in either CNGB3-/- or WT mice, but it increased the scotopic b-wave k (P < 0.01) in CNGB3-/- mice, indicating diminished scotopic sensitivity, and reduced the scotopic b-wave Vmax in WT mice (P < 0.05). No difference was found in ERG parameters between 1 or 2 µg CNTF. Fourteen days after CNTF injection the ERG changes in CNGB3-/- mice were lost. CONCLUSIONS: Intravitreal bolus CNTF protein caused a small and transient improvement of cone-mediated function in CNGB3-/- mice, whereas it reduced rod-mediated function. The increase in photopic OPs and the lack of changes in scotopic a-wave suggest a CNTF effect on the inner retina.


Subject(s)
Ciliary Neurotrophic Factor/administration & dosage , Color Vision Defects/drug therapy , Retinal Cone Photoreceptor Cells/drug effects , Animals , Color Vision Defects/physiopathology , Disease Models, Animal , Drug Implants , Electroretinography , Intravitreal Injections , Mice , Mice, Transgenic , Retinal Cone Photoreceptor Cells/physiology
14.
Cold Spring Harb Perspect Med ; 5(8): a017368, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26101206

ABSTRACT

Retinoschisis is an X-linked recessive genetic disease that leads to vision loss in males. X-linked retinoschisis (XLRS) typically affects young males; however, progressive vision loss continues throughout life. Although discovered in 1898 by Haas in two brothers, the underlying biology leading to blindness has become apparent only in the last 15 years with the advancement of human genetic analyses, generation of XLRS animal models, and the development of ocular monitoring methods such as the electroretinogram and optical coherence tomography. It is now recognized that retinoschisis results from cyst formations within the retinal layers that interrupt normal visual neurosignaling and compromise structural integrity. Mutations in the human retinoschisin gene have been correlated with disease severity of the human XLRS phenotype. Introduction of a normal human retinoschisin cDNA into retinoschisin knockout mice restores retinal structure and improves neural function, providing proof-of-concept that gene replacement therapy is a plausible treatment for XLRS.


Subject(s)
Genetic Predisposition to Disease , Genetic Therapy/methods , Retinoschisis/genetics , Retinoschisis/therapy , Animals , Disease Models, Animal , Disease Progression , Electroretinography/methods , Female , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Genetics, Medical/methods , Humans , Male , Mice , Mice, Knockout , Mutation , Phenotype , Prognosis , Prospective Studies , Rare Diseases , Risk Assessment , Treatment Outcome
15.
Exp Hematol ; 43(7): 514-23.e1-2, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25951974

ABSTRACT

Inactivating mutations in IKZF1, the gene that encodes the transcription factor IKAROS, are recurrent in poor-prognosis human B-cell leukemias, in which these mutations co-exist with BCR-ABL1 or other genetic changes that activate similar intracellular signaling pathways. However, little is known about the mechanism(s) by which loss of IKAROS activity may co-operate with BCR-ABL1 to transform lymphoid cells. To investigate this question, we used expression of a dominant-negative isoform of IKAROS (IK6) to suppress endogenous IKAROS activity in the interleukin-3 (IL-3)-dependent mouse pro-B BA/F3 cell line and in an IL-3-independent BCR-ABL1(+) derivative. We then used intracellular phospho-flow cytometry to assess the effects of BCR-ABL1 and IK6, alone and in combination, on the signaling state of the cells before and after their stimulation with IL-3. BCR-ABL1 and IK6 each produced a constitutively activated signaling phenotype and also enhanced the signaling responses of BA/F3 cells to IL-3. These effects, however, were neither equivalent nor additive, and IK6 alone was insufficient to confer the IL-3-independent growth characteristic of BCR-ABL1(+) BA/F3 cells. In addition to its effects on lymphoid cells, IK6 also induced constitutively activated signaling in a subset of myeloid leukemia cell lines. Together, these studies indicate an ability of IK6 to enhance intracellular signaling in both lymphoid and myeloid cells, but not to synergize with BCR-ABL1 in this model system.


Subject(s)
B-Lymphocytes/metabolism , Ikaros Transcription Factor/physiology , Interleukin-3/agonists , Signal Transduction/genetics , Animals , B-Lymphocytes/drug effects , Cell Division , Cell Line, Transformed , Cell Lineage , Cell Survival/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/physiology , Genes, Dominant , Humans , Ikaros Transcription Factor/antagonists & inhibitors , Ikaros Transcription Factor/genetics , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , Mutation , Myeloid Cells/cytology , Myeloid Cells/metabolism , Phenotype , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Transduction, Genetic
16.
Stem Cell Reports ; 3(5): 841-57, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418728

ABSTRACT

Disrupted IKAROS activity is a recurrent feature of some human leukemias, but effects on normal human hematopoietic cells are largely unknown. Here, we used lentivirally mediated expression of a dominant-negative isoform of IKAROS (IK6) to block normal IKAROS activity in primitive human cord blood cells and their progeny. This produced a marked (10-fold) increase in serially transplantable multipotent IK6(+) cells as well as increased outputs of normally differentiating B cells and granulocytes in transplanted immunodeficient mice, without producing leukemia. Accompanying T/natural killer (NK) cell outputs were unaltered, and erythroid and platelet production was reduced. Mechanistically, IK6 specifically increased human granulopoietic progenitor sensitivity to two growth factors and activated CREB and its targets (c-FOS and Cyclin B1). In more primitive human cells, IK6 prematurely initiated a B cell transcriptional program without affecting the hematopoietic stem cell-associated gene expression profile. Some of these effects were species specific, thus identifying novel roles of IKAROS in regulating normal human hematopoietic cells.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Ikaros Transcription Factor/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Blotting, Western , Cell Differentiation , Cells, Cultured , Fetal Blood/cytology , Gene Expression Profiling , Granulocytes/cytology , Granulocytes/metabolism , HeLa Cells , Hematopoietic Stem Cell Transplantation/methods , Humans , Ikaros Transcription Factor/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Confocal , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transplantation, Heterologous
17.
Invest Ophthalmol Vis Sci ; 55(10): 6301-8, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25205868

ABSTRACT

PURPOSE: Ciliary neurotrophic factor (CNTF) protects rod photoreceptors from retinal degenerative disease in multiple nonhuman models. Thus far, CNTF has failed to demonstrate rod protection in trials for human retinitis pigmentosa. Recently, CNTF was found to improve cone photoreceptor function in a canine CNGB3 achromatopsia model. This study explores whether this finding translates to humans with CNGB3 achromatopsia. METHODS: A five-subject, open-label Phase I/II study was initiated by implanting intraocular microcapsules releasing CNTF (nominally 20 ng/d) into one eye each of CNGB3 achromat participants. Fellow eyes served as untreated controls. Subjects were followed for 1 year. RESULTS: Pupil constriction in treated eyes gave evidence of intraocular CNTF release. Additionally, scotopic ERG responses were reduced, and dark-adapted psychophysical absolute thresholds were increased, attributable to diminished rod or rod pathway activity. Optical coherence tomography revealed that the cone-rich fovea underwent structural changes as the foveal hyporeflective zone (HRZ) became diminished in CNTF-treated eyes. No objectively measurable enhancement of cone function was found by assessments of visual acuity, mesopic increment sensitivity threshold, or the photopic ERG. Careful measurements of color hue discrimination showed no change. Nonetheless, subjects reported beneficial changes of visual function in the treated eyes, including reduced light sensitivity and aversion to bright light, which may trace to decreased effective ambient light from the pupillary constriction; further they noted slowed adaptation to darkness, consistent with CNTF action on rod photoreceptors. CONCLUSIONS: Ciliary neurotrophic factor did not measurably enhance cone function, which reveals a species difference between human and canine CNGB3 cones in response to CNTF. (ClinicalTrials.gov number, NCT01648452.).


Subject(s)
Ciliary Neurotrophic Factor/administration & dosage , Color Vision Defects/drug therapy , Cyclic Nucleotide-Gated Cation Channels/metabolism , Retinal Rod Photoreceptor Cells/physiology , Adult , Capsules , Color Vision Defects/metabolism , Color Vision Defects/physiopathology , Dark Adaptation , Drug Implants , Electroretinography , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies , Retinal Rod Photoreceptor Cells/drug effects , Time Factors , Tomography, Optical Coherence , Young Adult
18.
Hum Gene Ther Clin Dev ; 25(4): 202-11, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25211193

ABSTRACT

X-linked retinoschisis (XLRS) is a retinal disease caused by mutations in the gene encoding the protein retinoschisin (RS1) and one of the most common causes of macular degeneration in young men. Currently, no FDA-approved treatments are available for XLRS and a replacement gene therapy could provide a promising strategy. We have developed a novel gene therapy approach for XLRS, based on the administration of AAV8-scRS/IRBPhRS, an adeno-associated viral vector coding the human RS1 protein, via the intravitreal route. On the basis of our prior study in an Rs1-KO mouse, this construct transduces efficiently all the retinal layers, resulting in an RS1 expression similar to that observed in the wild-type and improving retinal structure and function. In support of a clinical trial, we carried out a study to evaluate the ocular safety of intravitreal administration of AAV8-scRS/IRBPhRS into 39 New Zealand White rabbits. Two dose levels of vector, 2e(10) and 2e(11) vector genomes per eye (vg/eye), were tested and ocular inflammation was monitored over a 12-week period by serial ophthalmological and histopathological analysis. A mild ocular inflammatory reaction, consisting mainly of vitreous infiltrates, was observed within 4 weeks from injection, in both 2e(10) and 2e(11) vg/eye groups and was likely driven by the AAV8 capsid. At 12-week follow-up, ophthalmological examination revealed no clinical signs of vitreitis in either of the dose groups. However, while vitreous inflammatory infiltrate was significantly reduced in the 2e(10) vg/eye group at 12 weeks, some rabbits in the higher dose group still showed persistence of inflammatory cells, histologically. In conclusion, intravitreal administration of AAV8-scRS/IRBPhRS into the rabbit eye produces a mild and transient intraocular inflammation that resolves, at a 2e(10) vg/eye dose, within 3 months, and does not cause irreversible tissue damages. These data support the initiation of a clinical trial of intravitreal administration of AAV8-scRS/IRBPhRS in XLRS patients.


Subject(s)
DNA, Recombinant/adverse effects , Dependovirus/genetics , Eye Proteins/genetics , Genetic Therapy , Genetic Vectors/adverse effects , Retinoschisis/therapy , Animals , DNA, Recombinant/administration & dosage , DNA, Recombinant/genetics , Dependovirus/metabolism , Eye Proteins/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Intravitreal Injections , Rabbits
19.
PLoS One ; 9(9): e108071, 2014.
Article in English | MEDLINE | ID: mdl-25232844

ABSTRACT

PURPOSE: To evaluate localization and transgene expression from adenoviral vector of serotypes 5, 35, and 28, ± an RGD motif in the fiber following intravitreal or subretinal administration. METHODS: Ocular transduction by adenoviral vector serotypes ± RGD was studied in the eyes of mice receiving an intravitreous or subretinal injection. Each serotype expressed a CMV-GFP expression cassette and histological sections of eyes were examined. Transgene expression levels were examined using luciferase (Luc) regulated by the CMV promoter. RESULTS: GFP localization studies revealed that serotypes 5 and 28 given intravitreously transduced corneal endothelial, trabecular, and iris cells. Intravitreous delivery of the unmodified Ad35 serotype transduced only trabecular meshwork cells, but, the modification of the RGD motif into the fiber of the Ad35 viral vector base expanded transduction to corneal endothelial and iris cells. Incorporation of the RGD motif into the fiber knob with deletion of RGD from the penton base did not affect the transduction ability of the Ad5 vector base. Subretinal studies showed that RGD in the Ad5 knob shifted transduction from RPE cells to photoreceptor cells. Using a CMV-Luc expression cassette, intravitreous delivery of all the tested vectors, such as Ad5-, Ad35- and Ad28- resulted in an initial rapid induction of luciferase activity that thereafter declined. Subretinal administration of vectors showed a marked difference in transgene activity. Ad35-Luc gene expression peaked at 7 days and remained elevated for 6 months. Ad28-Luc expression was high after 1 day and remained sustained for one month. CONCLUSIONS: Different adenoviral vector serotypes ± modifications transduce different cells within the eye. Transgene expression can be brief or extended and is serotype and delivery route dependent. Thus, adenoviral vectors provide a versatile platform for the delivery of therapeutic agents for ocular diseases.


Subject(s)
Adenoviridae/genetics , Oligopeptides/genetics , Animals , Female , Gene Expression , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Injections, Intraocular , Mice, Inbred C57BL , Transduction, Genetic
20.
Mol Ther ; 16(12): 1986-94, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18827806

ABSTRACT

Ocular neovascularization, the growth of abnormal blood vessels in the eye, is a factor shared by the most common blinding diseases in developed countries. Pigment epithelium-derived factor (PEDF) is a potent antiangiogenic and neuroprotective protein that is normally produced in the eye. When delivered via an adenovector, PEDF can block the growth of new blood vessels and trigger the selective regression of abnormal vessels in animal models of ocular disease. Because of the absence of adenoviral genes, high-capacity (HC) adenovectors offer the potential for persistent transgene expression and enhanced tolerability. We have assessed the durability of PEDF expression and the induction of ocular inflammation following delivery of a PEDF-expressing HC adenovector compared to earlier generation vectors. The HC vector mediated prolonged PEDF expression in tissue-cultured pigmented epithelial cells and when delivered by intravitreal injection into the mouse eye. Delivery of first-generation adenovectors resulted in a dose-dependent increase in cytokine/chemokine gene expression, which correlated with the infiltration of inflammatory cells in the eye. In comparison, the levels of inflammatory gene expression and the intraocular infiltrate were substantially reduced following delivery of the HC vector. These results support the development of the HC adenovector gene delivery system for ocular disease.


Subject(s)
Adenoviridae/genetics , Eye Proteins/metabolism , Eye/metabolism , Gene Expression , Genetic Vectors/genetics , Nerve Growth Factors/metabolism , Serpins/metabolism , Administration, Intravesical , Animals , Chemokines/genetics , Eye Diseases/genetics , Eye Diseases/metabolism , Eye Diseases/pathology , Eye Proteins/genetics , Female , Gene Deletion , Gene Expression Regulation , Genetic Vectors/administration & dosage , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Kinetics , Mice , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Serpins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...