Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell ; 187(10): 2521-2535.e21, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38697107

ABSTRACT

Cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Here, we create "onion-like" multi-lamellar RNA lipid particle aggregates (LPAs) to substantially enhance the payload packaging and immunogenicity of tumor mRNA antigens. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for Toll-like receptor engagement in immune cells, systemically administered RNA-LPAs activate RIG-I in stromal cells, eliciting massive cytokine/chemokine response and dendritic cell/lymphocyte trafficking that provokes cancer immunogenicity and mediates rejection of both early- and late-stage murine tumor models. In client-owned canines with terminal gliomas, RNA-LPAs improved survivorship and reprogrammed the TME, which became "hot" within days of a single infusion. In a first-in-human trial, RNA-LPAs elicited rapid cytokine/chemokine release, immune activation/trafficking, tissue-confirmed pseudoprogression, and glioma-specific immune responses in glioblastoma patients. These data support RNA-LPAs as a new technology that simultaneously reprograms the TME while eliciting rapid and enduring cancer immunotherapy.


Subject(s)
Immunotherapy , Lipids , RNA , Tumor Microenvironment , Animals , Dogs , Female , Humans , Mice , Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glioblastoma/therapy , Glioblastoma/immunology , Glioma/therapy , Glioma/immunology , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/therapy , Neoplasms/immunology , RNA/chemistry , RNA/therapeutic use , RNA, Messenger/metabolism , RNA, Messenger/genetics , Lipids/chemistry
2.
medRxiv ; 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36993772

ABSTRACT

Messenger RNA (mRNA) has emerged as a remarkable tool for COVID-19 prevention but its use for induction of therapeutic cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Herein, we develop a facile approach for substantially enhancing immunogenicity of tumor-derived mRNA in lipid-particle (LP) delivery systems. By using mRNA as a molecular bridge with ultrapure liposomes and foregoing helper lipids, we promote the formation of 'onion-like' multi-lamellar RNA-LP aggregates (LPA). Intravenous administration of RNA-LPAs mimics infectious emboli and elicits massive DC/T cell mobilization into lymphoid tissues provoking cancer immunogenicity and mediating rejection of both early and late-stage murine tumor models. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for toll-like receptor engagement, RNA-LPAs stimulate intracellular pathogen recognition receptors (RIG-I) and reprogram the TME thus enabling therapeutic T cell activity. RNA-LPAs were safe in acute/chronic murine GLP toxicology studies and immunologically active in client-owned canines with terminal gliomas. In an early phase first-in-human trial for patients with glioblastoma, we show that RNA-LPAs encoding for tumor-associated antigens elicit rapid induction of pro-inflammatory cytokines, mobilization/activation of monocytes and lymphocytes, and expansion of antigen-specific T cell immunity. These data support the use of RNA-LPAs as novel tools to elicit and sustain immune responses against poorly immunogenic tumors.

3.
Mol Neurobiol ; 59(9): 5750-5765, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35781633

ABSTRACT

Changes in the transcription factor (TF) expression are critical for brain development, and they may also underlie neurodevelopmental disorders. Indeed, T-box brain1 (Tbr1) is a TF crucial for the formation of neocortical layer VI, and mutations and microdeletions in that gene are associated with malformations in the human cerebral cortex, alterations that accompany autism spectrum disorder (ASD). Interestingly, Tbr1 upregulation has also been related to the occurrence of ASD-like symptoms, although limited studies have addressed the effect of increased Tbr1 levels during neocortical development. Here, we analysed the impact of Tbr1 misexpression in mouse neural progenitor cells (NPCs) at embryonic day 14.5 (E14.5), when they mainly generate neuronal layers II-IV. By E18.5, cells accumulated in the intermediate zone and in the deep cortical layers, whereas they became less abundant in the upper cortical layers. In accordance with this, the proportion of Sox5+ cells in layers V-VI increased, while that of Cux1+ cells in layers II-IV decreased. On postnatal day 7, fewer defects in migration were evident, although a higher proportion of Sox5+ cells were seen in the upper and deep layers. The abnormal neuronal migration could be partially due to the altered multipolar-bipolar neuron morphologies induced by Tbr1 misexpression, which also reduced dendrite growth and branching, and disrupted the corpus callosum. Our results indicate that Tbr1 misexpression in cortical NPCs delays or disrupts neuronal migration, neuronal specification, dendrite development and the formation of the callosal tract. Hence, genetic changes that provoke ectopic Tbr1 upregulation during development could provoke cortical brain malformations.


Subject(s)
Autism Spectrum Disorder , Neocortex , Animals , Autism Spectrum Disorder/genetics , Cerebral Cortex/metabolism , Humans , Mice , Neocortex/metabolism , Neurogenesis/genetics , Neurons/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
4.
Cells ; 11(12)2022 06 20.
Article in English | MEDLINE | ID: mdl-35741103

ABSTRACT

The promising outcomes of chimeric antigen receptor (CAR) T cell therapy in hematologic malignancies potentiates its capability in the fight against many cancers. Nevertheless, this immunotherapy modality needs significant improvements for the treatment of solid tumors. Researchers have incrementally identified limitations and constantly pursued better CAR designs. However, even if CAR T cells are armed with optimal killer functions, they must overcome and survive suppressive barriers imposed by the tumor microenvironment (TME). In this review, we will discuss in detail the important role of TME in CAR T cell trafficking and how the intrinsic barriers contribute to an immunosuppressive phenotype and cancer progression. It is of critical importance that preclinical models can closely recapitulate the in vivo TME to better predict CAR T activity. Animal models have contributed immensely to our understanding of human diseases, but the intensive care for the animals and unreliable representation of human biology suggest in vivo models cannot be the sole approach to CAR T cell therapy. On the other hand, in vitro models for CAR T cytotoxic assessment offer valuable insights to mechanistic studies at the single cell level, but they often lack in vivo complexities, inter-individual heterogeneity, or physiologically relevant spatial dimension. Understanding the advantages and limitations of preclinical models and their applications would enable more reliable prediction of better clinical outcomes.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Animals , Cell Movement , Immunotherapy, Adoptive/methods , Neoplasms/pathology , T-Lymphocytes , Tumor Microenvironment
5.
J Neurooncol ; 151(1): 29-39, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32757093

ABSTRACT

INTRODUCTION: Brain tumors remain especially challenging to treat due to the presence of the blood-brain barrier. The unique biophysical properties of nanomaterials enable access to the tumor environment with minimally invasive injection methods such as intranasal and systemic delivery. METHODS: In this review, we will discuss approaches taken in NP delivery to brain tumors in preclinical neuro-oncology studies and ongoing clinical studies. RESULTS: Despite recent development of many promising nanoparticle systems to modulate immunologic function in the preclinical realm, clinical work with nanoparticles in malignant brain tumors has largely focused on imaging, chemotherapy, thermotherapy and radiation. CONCLUSION: Review of early preclinical studies and clinical trials provides foundational safety, feasibility and toxicology data that can usher a new wave of nanotherapeutics in application of immunotherapy and translational oncology for patients with brain tumors.


Subject(s)
Brain Neoplasms , Nanoparticles , Adjuvants, Immunologic/therapeutic use , Blood-Brain Barrier , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Drug Delivery Systems , Humans , Immunologic Factors/therapeutic use
6.
ACS Nano ; 13(12): 13884-13898, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31730332

ABSTRACT

Cancer vaccines initiate antitumor responses in a subset of patients, but the lack of clinically meaningful biomarkers to predict treatment response limits their development. Here, we design multifunctional RNA-loaded magnetic liposomes to initiate potent antitumor immunity and function as an early biomarker of treatment response. These particles activate dendritic cells (DCs) more effectively than electroporation, leading to superior inhibition of tumor growth in treatment models. Inclusion of iron oxide enhances DC transfection and enables tracking of DC migration with magnetic resonance imaging (MRI). We show that T2*-weighted MRI intensity in lymph nodes is a strong correlation of DC trafficking and is an early predictor of antitumor response. In preclinical tumor models, MRI-predicted "responders" identified 2 days after vaccination had significantly smaller tumors 2-5 weeks after treatment and lived 73% longer than MRI-predicted "nonresponders". These studies therefore provide a simple, scalable nanoparticle formulation to generate robust antitumor immune responses and predict individual treatment outcome with MRI.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/metabolism , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Animals , Biomarkers, Tumor/metabolism , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Movement/drug effects , Cell Tracking , Dendritic Cells/drug effects , Electroporation , Ferric Compounds/chemistry , Magnetite Nanoparticles/ultrastructure , Mice, Inbred C57BL , Transfection
7.
J Hematol Oncol ; 12(1): 78, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31311607

ABSTRACT

While promising, immunotherapy has yet to be fully unlocked for the preponderance of cancers where conventional chemoradiation reigns. This remains particularly evident in pediatric sarcomas where standard of care has not appreciably changed in decades. Importantly, pediatric bone sarcomas, like osteosarcoma and Ewing's sarcoma, possess unique tumor microenvironments driven by distinct molecular features, as do rhabdomyosarcomas and soft tissue sarcomas. A better understanding of each malignancy's biology, heterogeneity, and tumor microenvironment may lend new insights toward immunotherapeutic targets in novel platform technologies for cancer vaccines and adoptive cellular therapy. These advances may pave the way toward new treatments requisite for pediatric sarcomas and patients in need of new therapies.


Subject(s)
Immunotherapy/methods , Sarcoma/drug therapy , Adolescent , Child , Child, Preschool , Female , Humans , Male , Tumor Microenvironment
8.
Int J Mol Sci ; 19(10)2018 Sep 23.
Article in English | MEDLINE | ID: mdl-30249040

ABSTRACT

Cancer vaccines may be harnessed to incite immunity against poorly immunogenic tumors, however they have failed in therapeutic settings. Poor antigenicity coupled with systemic and intratumoral immune suppression have been significant drawbacks. RNA encoding for tumor associated or specific epitopes can serve as a more immunogenic and expeditious trigger of anti-tumor immunity. RNA stimulates innate immunity through toll like receptor stimulation producing type I interferon, and it mediates potent adaptive responses. Since RNA is inherently unstable, delivery systems have been developed to protect and deliver it to intended targets in vivo. In this review, we discuss liposomes as RNA delivery vehicles and their role as cancer vaccines.


Subject(s)
Cancer Vaccines/administration & dosage , Immunotherapy , Liposomes/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , RNA/administration & dosage , Animals , Cancer Vaccines/chemistry , Drug Delivery Systems , Humans , Liposomes/chemistry , RNA/chemistry
9.
Nano Lett ; 18(10): 6195-6206, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30259750

ABSTRACT

Translation of nanoparticles (NPs) into human clinical trials for patients with refractory cancers has lagged due to unknown biologic reactivities of novel NP designs. To overcome these limitations, simple well-characterized mRNA lipid-NPs have been developed as cancer immunotherapeutic vaccines. While the preponderance of RNA lipid-NPs encoding for tumor-associated antigens or neoepitopes have been designed to target lymphoid organs, they remain encumbered by the profound intratumoral and systemic immunosuppression that may stymie an activated T cell response. Herein, we show that systemic localization of untargeted tumor RNA (derived from whole transcriptome) encapsulated in lipid-NPs, with excess positive charge, primes the peripheral and intratumoral milieu for response to immunotherapy. In immunologically resistant tumor models, these RNA-NPs activate the preponderance of systemic and intratumoral myeloid cells (characterized by coexpression of PD-L1 and CD86). Addition of immune checkpoint inhibitors (ICIs) (to animals primed with RNA-NPs) augments peripheral/intratumoral PD-1+CD8+ cells and mediates synergistic antitumor efficacy in settings where ICIs alone do not confer therapeutic benefit. These synergistic effects are mediated by type I interferon released from plasmacytoid dendritic cells (pDCs). In translational studies, personalized mRNA-NPs were safe and active in a client-owned canine with a spontaneous malignant glioma. In summary, we demonstrate widespread immune activation from tumor loaded RNA-NPs concomitant with inducible PD-L1 expression that can be therapeutically exploited. While immunotherapy remains effective for only a subset of cancer patients, combination therapy with systemic immunomodulating RNA-NPs may broaden its therapeutic potency.


Subject(s)
Glioma/drug therapy , Immunotherapy , Lipids/administration & dosage , Nanoparticles/administration & dosage , Precision Medicine , Animals , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/genetics , B7-2 Antigen/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Disease Models, Animal , Dogs , Glioma/immunology , Glioma/pathology , Glioma/veterinary , Humans , Lipids/chemistry , Lipids/immunology , Lymphocyte Activation/immunology , Nanoparticles/chemistry , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Transcriptome/genetics
10.
Neuroscience ; 377: 174-183, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29526688

ABSTRACT

Phospholipase D2 (PLD2), an enzyme involved in vesicle trafficking and membrane signaling, interacts with α-synuclein, a protein known to contribute in the development of Parkinson disease (PD). We previously reported that PLD2 overexpression in rat substantia nigra pars compacta (SNc) causes a rapid neurodegeneration of dopamine neurons, and that α-synuclein suppresses PLD2-induced nigral degeneration (Gorbatyuk et al., 2010). Here, we report that PLD2 toxicity is due to its lipase activity. Overexpression of a catalytically inactive mutant (K758R) of PLD2 prevents the loss of dopaminergic neurons in the SNc and does not show signs of toxicity after 10 weeks of overexpression. Further, mutant K758R does not affect dopamine levels in the striatum. In contrast, mutants that prevent PLD2 interaction with dynamin or growth factor receptor bound protein 2 (Grb2) but retained lipase activity, continued to show rapid neurodegeneration. These findings suggest that neither the interaction of PLD2 with dynamin, which has a role in vesicle trafficking, nor the PLD2 interaction with Grb2, which has multiple roles in cell cycle control, chemotaxis and activation of tyrosine kinase complexes, are the primary cause of neurodegeneration. Instead, the synthesis of phosphatidic acid (the product of PLD2), which is a second messenger in multiple cellular pathways, appears to be the key to PLD2 induced neurodegeneration. The fact that α-synuclein is a regulator of PLD2 activity suggests that regulation of PLD2 activity could be important in the progression of PD.


Subject(s)
Nerve Degeneration/enzymology , Parkinsonian Disorders/enzymology , Pars Compacta/enzymology , Phospholipase D/metabolism , Animals , Dynamins/metabolism , GRB2 Adaptor Protein/metabolism , Gene Expression , HEK293 Cells , Humans , Mutation , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/pathology , Parkinsonian Disorders/pathology , Pars Compacta/pathology , Phospholipase D/genetics , Rats , Tyrosine 3-Monooxygenase/metabolism
11.
Mol Cell ; 68(3): 479-490.e5, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29056323

ABSTRACT

Transcription of expanded microsatellite repeats is associated with multiple human diseases, including myotonic dystrophy, Fuchs endothelial corneal dystrophy, and C9orf72-ALS/FTD. Reducing production of RNA and proteins arising from these expanded loci holds therapeutic benefit. Here, we tested the hypothesis that deactivated Cas9 enzyme impedes transcription across expanded microsatellites. We observed a repeat length-, PAM-, and strand-dependent reduction of repeat-containing RNAs upon targeting dCas9 directly to repeat sequences; targeting the non-template strand was more effective. Aberrant splicing patterns were rescued in DM1 cells, and production of RAN peptides characteristic of DM1, DM2, and C9orf72-ALS/FTD cells was drastically decreased. Systemic delivery of dCas9/gRNA by adeno-associated virus led to reductions in pathological RNA foci, rescue of chloride channel 1 protein expression, and decreased myotonia. These observations suggest that transcription of microsatellite repeat-containing RNAs is more sensitive to perturbation than transcription of other RNAs, indicating potentially viable strategies for therapeutic intervention.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Endonucleases/metabolism , Genetic Therapy/methods , Microsatellite Repeats , Myotonic Dystrophy/therapy , Transcription, Genetic , Alternative Splicing , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Dependovirus/genetics , Disease Models, Animal , Down-Regulation , Enzyme Activation , Female , Genetic Vectors , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Transgenic , Myoblasts/metabolism , Myoblasts/pathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA, Guide, Kinetoplastida/biosynthesis , RNA, Guide, Kinetoplastida/genetics , Transduction, Genetic , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
12.
Mol Ther ; 25(12): 2661-2675, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-28890324

ABSTRACT

The major drawback of the Baculovirus/Sf9 system for recombinant adeno-associated viral (rAAV) manufacturing is that most of the Bac-derived rAAV vector serotypes, with few exceptions, demonstrate altered capsid compositions and lower biological potencies. Here, we describe a new insect cell-based production platform utilizing attenuated Kozak sequence and a leaky ribosome scanning to achieve a serotype-specific modulation of AAV capsid proteins stoichiometry. By way of example, rAAV5 and rAAV9 were produced and comprehensively characterized side by side with HEK293-derived vectors. A mass spectrometry analysis documented a 3-fold increase in both viral protein (VP)1 and VP2 capsid protein content compared with human cell-derived vectors. Furthermore, we conducted an extensive analysis of encapsidated single-stranded viral DNA using next-generation sequencing and show a 6-fold reduction in collaterally packaged contaminating DNA for rAAV5 produced in insect cells. Consequently, the re-designed rAAVs demonstrated significantly higher biological potencies, even in a comparison with HEK293-manufactured rAAVs mediating, in the case of rAAV5, 4-fold higher transduction of brain tissues in mice. Thus, the described system yields rAAV vectors of superior infectivity and higher genetic identity providing a scalable platform for good manufacturing practice (GMP)-grade vector production.


Subject(s)
Cell Culture Techniques , Dependovirus/genetics , Genetic Vectors/genetics , Virus Replication , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Dependovirus/classification , Dependovirus/physiology , Gene Expression , Gene Order , Genes, Reporter , HEK293 Cells , Humans , Mice , Sf9 Cells , Tissue Distribution , Transduction, Genetic , Viral Load
13.
Article in English | MEDLINE | ID: mdl-27999773

ABSTRACT

Graphene, graphene-based nanomaterials (GBNs), and carbon nanotubes (CNTs) are being investigated as potential substrates for the growth of neural cells. However, in most in vitro studies, the cells were seeded on these materials coated with various proteins implying that the observed effects on the cells could not solely be attributed to the GBN and CNT properties. Here, we studied the biocompatibility of uncoated thermally reduced graphene (TRG) and poly(vinylidene fluoride) (PVDF) membranes loaded with multi-walled CNTs (MWCNTs) using neural stem cells isolated from the adult mouse olfactory bulb (termed aOBSCs). When aOBSCs were induced to differentiate on coverslips treated with TRG or control materials (polyethyleneimine-PEI and polyornithine plus fibronectin-PLO/F) in a serum-free medium, neurons, astrocytes, and oligodendrocytes were generated in all conditions, indicating that TRG permits the multi-lineage differentiation of aOBSCs. However, the total number of cells was reduced on both PEI and TRG. In a serum-containing medium, aOBSC-derived neurons and oligodendrocytes grown on TRG were more numerous than in controls; the neurons developed synaptic boutons and oligodendrocytes were more branched. In contrast, neurons growing on PVDF membranes had reduced neurite branching, and on MWCNTs-loaded membranes oligodendrocytes were lower in numbers than in controls. Overall, these findings indicate that uncoated TRG may be biocompatible with the generation, differentiation, and maturation of aOBSC-derived neurons and glial cells, implying a potential use for TRG to study functional neuronal networks.

14.
Neurotox Res ; 30(1): 14-31, 2016 07.
Article in English | MEDLINE | ID: mdl-26678495

ABSTRACT

NURR1 is an essential transcription factor for the differentiation, maturation, and maintenance of midbrain dopaminergic neurons (DA neurons) as it has been demonstrated using knock-out mice. DA neurons of the substantia nigra pars compacta degenerate in Parkinson's disease (PD) and mutations in the Nurr1 gene have been associated with this human disease. Thus, the study of NURR1 actions in vivo is fundamental to understand the mechanisms of neuron generation and degeneration in the dopaminergic system. Here, we present and discuss findings indicating that NURR1 is a valuable molecular tool for the in vitro generation of DA neurons which could be used for modeling and studying PD in cell culture and in transplantation approaches. Transduction of Nurr1 alone or in combination with other transcription factors such as Foxa2, Ngn2, Ascl1, and Pitx3, induces the generation of DA neurons, which upon transplantation have the capacity to survive and restore motor behavior in animal models of PD. We show that the survival of transplanted neurons is increased when the Nurr1-transduced olfactory bulb stem cells are treated with GDNF. The use of these and other factors with the induced pluripotent stem cell (iPSC)-based technology or the direct reprogramming of astrocytes or fibroblasts into human DA neurons has produced encouraging results for the study of the cellular and molecular mechanisms of neurodegeneration in PD and for the search of new treatments for this disease.


Subject(s)
Dopaminergic Neurons/physiology , Neurogenesis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Animals , Cells, Cultured , Humans , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Olfactory Bulb/cytology , Stem Cell Transplantation/methods , Stem Cells/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/physiology
15.
Mol Ther Methods Clin Dev ; 2: 15037, 2015.
Article in English | MEDLINE | ID: mdl-26491705

ABSTRACT

Crossing the blood-brain and the blood-cerebrospinal fluid barriers (BCSFB) is one of the fundamental challenges in the development of new therapeutic molecules for brain disorders because these barriers prevent entry of most drugs from the blood into the brain. However, some large molecules, like the protein transferrin, cross these barriers using a specific receptor that transports them into the brain. Based on this mechanism, we engineered a receptor/ligand system to overcome the brain barriers by combining the human transferrin receptor with the cohesin domain from Clostridium thermocellum, and we tested the hybrid receptor in the choroid plexus of the mouse brain with a dockerin ligand. By expressing our receptor in choroidal ependymocytes, which are part of the BCSFB, we found that our systemically administrated ligand was able to bind to the receptor and accumulate in ependymocytes, where some of the ligand was transported from the blood side to the brain side.

16.
Dev Neurobiol ; 75(8): 823-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25447275

ABSTRACT

The transcription factor Nurr1 is expressed in the mouse olfactory bulb (OB), although it remains unknown whether it influences the generation of dopaminergic neurons (DA) (DA neurons) in cells isolated from this brain region. We found that expressing Nurr1 in proliferating olfactory bulb stem cells (OBSCs) produces a marked inhibition of cell proliferation and the generation of immature neurons immunoreactive for tyrosine hydroxylase (TH) concomitant with marked upregulations of Th, Dat, Gad, and Fgfr2 transcripts. In long-term cultures, these cells develop neurochemical and synaptic markers of mature-like mesencephalic DA neurons, expressing GIRK2, VMAT2, DAT, calretinin, calbindin, synapsin-I, and SV2. Concurring with the increase in both Th and Gad expression, a subpopulation of induced cells was both TH- and GAD-immunoreactive indicating that they are dopaminergic-GABAergic neurons. Indeed, these cells could mature to express VGAT, suggesting they can uptake and store GABA in vesicles. Remarkably, the dopamine D1 receptor agonist SKF-38393 induced c-Fos in TH(+) cells and dopamine release was detected in these cultures under basal and KCl-evoked conditions. By contrast, cotransducing the Neurogenin2 and Nurr1 transcription factors produced a significant decrease in the number of TH-positive neurons. Our results indicate that Nurr1 overexpression in OBSCs induces the formation of two populations of mature dopaminergic neurons with features of the ventral mesencephalon or of the OB, capable of responding to functional dopaminergic stimuli and of releasing dopamine. They also suggest that the accumulation of Fgfr2 by Nurr1 in OBSCs may be involved in the generation of DA neurons.


Subject(s)
Dopaminergic Neurons/physiology , GABAergic Neurons/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Olfactory Bulb/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dopaminergic Neurons/drug effects , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/metabolism , GABAergic Neurons/drug effects , Mice, Inbred C57BL , Mitosis/drug effects , Mitosis/physiology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Olfactory Bulb/drug effects , Tyrosine 3-Monooxygenase/metabolism
17.
Mol Ther Methods Clin Dev ; 2: 15041, 2015.
Article in English | MEDLINE | ID: mdl-26793739

ABSTRACT

Biodistribution analysis is a key step in the evaluation of adeno-associated virus (AAV) capsid variants, whether natural isolates or produced by rational design or directed evolution. Indeed, when screening candidate vectors, accurate knowledge about which tissues are infected and how efficiently is essential. We describe the design, validation, and application of a new vector, pTR-UF50-BC, encoding a bioluminescent protein, a fluorescent protein and a DNA barcode, which can be used to visualize localization of transduction at the organism, organ, tissue, or cellular levels. In addition, by linking capsid variants to different barcoded versions of the vector and amplifying the barcode region from various tissue samples using barcoded primers, biodistribution of viral genomes can be analyzed with high accuracy and efficiency.

18.
PLoS One ; 7(1): e29799, 2012.
Article in English | MEDLINE | ID: mdl-22242181

ABSTRACT

The Genetic screened homeobox 2 (Gsx2) transcription factor is required for the development of olfactory bulb (OB) and striatal neurons, and for the regional specification of the embryonic telencephalon. Although Gsx2 is expressed abundantly by progenitor cells in the ventral telencephalon, its precise function in the generation of neurons from neural stem cells (NSCs) is not clear. Similarly, the role of Gsx2 in regulating the self-renewal and multipotentiality of NSCs has been little explored. Using retroviral vectors to express Gsx2, we have studied the effect of Gsx2 on the growth of NSCs isolated from the OB and ganglionic eminences (GE), as well as its influence on the proliferation and cell fate of progenitors in the postnatal mouse OB. Expression of Gsx2 reduces proliferation and the self-renewal capacity of NSCs, without significantly affecting cell death. Furthermore, Gsx2 overexpression decreases the differentiation of NSCs into neurons and glia, and it maintains the cells that do not differentiate as cycling progenitors. These effects were stronger in GESCs than in OBSCs, indicating that the actions of Gsx2 are cell-dependent. In vivo, Gsx2 produces a decrease in the number of Pax6+ cells and doublecortin+ neuroblasts, and an increase in Olig2+ cells. In summary, our findings show that Gsx2 inhibits the ability of NSCs to proliferate and self-renew, as well as the capacity of NSC-derived progenitors to differentiate, suggesting that this transcription factor regulates the quiescent and undifferentiated state of NSCs and progenitors. Furthermore, our data indicate that Gsx2 negatively regulates neurogenesis from postnatal progenitor cells.


Subject(s)
Cell Differentiation/genetics , Cell Lineage , Homeodomain Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Biomarkers/metabolism , Cell Aggregation , Cell Death , Cell Division , Cell Proliferation , Embryo, Mammalian/cytology , Genetic Vectors/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Neurons/cytology , Olfactory Bulb/cytology , Oligodendroglia/cytology , Retroviridae/genetics , Transduction, Genetic
19.
Mol Cell Neurosci ; 46(1): 108-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20807572

ABSTRACT

The T-box brain 1 (Tbr1) gene encodes a transcription factor necessary for the maintenance and/or differentiation of glutamatergic cells in the olfactory bulb (OB) and cortex, although its precise function in the development of glutamatergic neurons is not known. Furthermore, Tbr1 has not been reported to regulate the formation of glial cells. We show that Tbr1 is expressed during the initial stages in the generation of glutamatergic mitral neurons from dividing progenitors in the E12.5 mouse OB. Retroviral-mediated overexpression of Tbr1 in cultured embryonic and adult OB stem cells (OBSC) produces a marked increase in the number of TuJ1(+) neurons (including VGLUT1(+) glutamatergic and GABA(+) neurons) and O4(+) oligodendrocytes. Moreover, transduction of Tbr1 inhibits the production of GFAP(+) astrocytes from both cultured OBSC and dividing progenitor cells in vivo. These results show that the expression of Tbr1 in neural stem and progenitor cells prevents them from following an astrocyte fate during OB development. Our findings suggest that the transduction of Tbr1 into neural stem cells could be useful to increase the production of neurons and oligodendrocytes in studies of neuroregeneration.


Subject(s)
Astrocytes/physiology , DNA-Binding Proteins/metabolism , Neural Stem Cells/physiology , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Animals , Astrocytes/cytology , Cell Differentiation/physiology , Cell Proliferation , DNA-Binding Proteins/genetics , Glutamic Acid/metabolism , Mice , Neural Stem Cells/cytology , Neurons/cytology , Neurons/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Box Domain Proteins , gamma-Aminobutyric Acid/metabolism
20.
Neural Dev ; 5: 21, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20735826

ABSTRACT

BACKGROUND: Nolz1 is a zinc finger transcription factor whose expression is enriched in the lateral ganglionic eminence (LGE), although its function is still unknown. RESULTS: Here we analyze the role of Nolz1 during LGE development. We show that Nolz1 expression is high in proliferating neural progenitor cells (NPCs) of the LGE subventricular zone. In addition, low levels of Nolz1 are detected in the mantle zone, as well as in the adult striatum. Similarly, Nolz1 is highly expressed in proliferating LGE-derived NPC cultures, but its levels rapidly decrease upon cell differentiation, pointing to a role of Nolz1 in the control of NPC proliferation and/or differentiation. In agreement with this hypothesis, we find that Nolz1 over-expression promotes cell cycle exit of NPCs in neurosphere cultures and negatively regulates proliferation in telencephalic organotypic cultures. Within LGE primary cultures, Nolz1 over-expression promotes the acquisition of a neuronal phenotype, since it increases the number of ß-III tubulin (Tuj1)- and microtubule-associated protein (MAP)2-positive neurons, and inhibits astrocyte generation and/or differentiation. Retinoic acid (RA) is one of the most important morphogens involved in striatal neurogenesis, and regulates Nolz1 expression in different systems. Here we show that Nolz1 also responds to this morphogen in E12.5 LGE-derived cell cultures. However, Nolz1 expression is not regulated by RA in E14.5 LGE-derived cell cultures, nor is it affected during LGE development in mouse models that present decreased RA levels. Interestingly, we find that Gsx2, which is necessary for normal RA signaling during LGE development, is also required for Nolz1 expression, which is lost in Gsx2 knockout mice. These findings suggest that Nolz1 might act downstream of Gsx2 to regulate RA-induced neurogenesis. Keeping with this hypothesis, we show that Nolz1 induces the selective expression of the RA receptor (RAR)ß without altering RARα or RARγ. In addition, Nozl1 over-expression increases RA signaling since it stimulates the RA response element. This RA signaling is essential for Nolz1-induced neurogenesis, which is impaired in a RA-free environment or in the presence of a RAR inverse agonist. It has been proposed that Drosophila Gsx2 and Nolz1 homologues could cooperate with the transcriptional co-repressors Groucho-TLE to regulate cell proliferation. In agreement with this view, we show that Nolz1 could act in collaboration with TLE-4, as they are expressed at the same time in NPC cultures and during mouse development. CONCLUSIONS: Nolz1 promotes RA signaling in the LGE, contributing to the striatal neurogenesis during development.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Corpus Striatum/cytology , Gene Expression Regulation, Developmental/drug effects , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Nuclear Proteins/metabolism , Signal Transduction/physiology , Tretinoin/pharmacology , Animals , Carrier Proteins/genetics , Cell Count , Cell Proliferation , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/physiology , Geniculate Bodies/embryology , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred CBA , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Neuroglia/drug effects , Neuroglia/physiology , Neurons/physiology , Nuclear Proteins/genetics , Pregnancy , Retinal Dehydrogenase/deficiency , Signal Transduction/genetics , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...