Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell ; 185(10): 1694-1708.e19, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35447074

ABSTRACT

Immunotherapy is a promising treatment for triple-negative breast cancer (TNBC), but patients relapse, highlighting the need to understand the mechanisms of resistance. We discovered that in primary breast cancer, tumor cells that resist T cell attack are quiescent. Quiescent cancer cells (QCCs) form clusters with reduced immune infiltration. They also display superior tumorigenic capacity and higher expression of chemotherapy resistance and stemness genes. We adapted single-cell RNA-sequencing with precise spatial resolution to profile infiltrating cells inside and outside the QCC niche. This transcriptomic analysis revealed hypoxia-induced programs and identified more exhausted T cells, tumor-protective fibroblasts, and dysfunctional dendritic cells inside clusters of QCCs. This uncovered differential phenotypes in infiltrating cells based on their intra-tumor location. Thus, QCCs constitute immunotherapy-resistant reservoirs by orchestrating a local hypoxic immune-suppressive milieu that blocks T cell function. Eliminating QCCs holds the promise to counteract immunotherapy resistance and prevent disease recurrence in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Immunosuppressive Agents/therapeutic use , Immunotherapy , Neoplasm Recurrence, Local , T-Lymphocytes/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
2.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34608934

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of the CAG repeats in the huntingtin gene (HTT). Although HD has been shown to have a developmental component, how early during human embryogenesis the HTT-CAG expansion can cause embryonic defects remains unknown. Here, we demonstrate a specific and highly reproducible CAG length-dependent phenotypic signature in a synthetic model for human gastrulation derived from human embryonic stem cells (hESCs). Specifically, we observed a reduction in the extension of the ectodermal compartment that is associated with enhanced activin signaling. Surprisingly, rather than a cell-autonomous effect, tracking the dynamics of TGFß signaling demonstrated that HTT-CAG expansion perturbs the spatial restriction of activin response. This is due to defects in the apicobasal polarization in the context of the polarized epithelium of the 2D gastruloid, leading to ectopic subcellular localization of TGFß receptors. This work refines the earliest developmental window for the prodromal phase of HD to the first 2 weeks of human development, as modeled by our 2D gastruloids.


Subject(s)
Cell Lineage , Cell Polarity , Germ Layers/metabolism , Human Embryonic Stem Cells/metabolism , Huntingtin Protein/metabolism , Activins/metabolism , Animals , Cell Line , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Germ Layers/cytology , Germ Layers/embryology , Human Embryonic Stem Cells/cytology , Humans , Huntingtin Protein/genetics , Mice , Signal Transduction , Transforming Growth Factor beta/metabolism , Trinucleotide Repeat Expansion
3.
Nat Cell Biol ; 21(7): 900-910, 2019 07.
Article in English | MEDLINE | ID: mdl-31263269

ABSTRACT

Breaking the anterior-posterior symmetry in mammals occurs at gastrulation. Much of the signalling network underlying this process has been elucidated in the mouse; however, there is no direct molecular evidence of events driving axis formation in humans. Here, we use human embryonic stem cells to generate an in vitro three-dimensional model of a human epiblast whose size, cell polarity and gene expression are similar to a day 10 human epiblast. A defined dose of BMP4 spontaneously breaks axial symmetry, and induces markers of the primitive streak and epithelial-to-mesenchymal transition. We show that WNT signalling and its inhibitor DKK1 play key roles in this process downstream of BMP4. Our work demonstrates that a model human epiblast can break axial symmetry despite the absence of asymmetry in the initial signal and of extra-embryonic tissues or maternal cues. Our three-dimensional model is an assay for the molecular events underlying human axial symmetry breaking.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Gene Expression Regulation, Developmental/physiology , Germ Layers/metabolism , Primitive Streak/metabolism , Tissue Culture Techniques , Cell Polarity/physiology , Epithelial-Mesenchymal Transition , Gastrulation/physiology , Humans , Primitive Streak/embryology , Signal Transduction/physiology
4.
Elife ; 72018 10 12.
Article in English | MEDLINE | ID: mdl-30311909

ABSTRACT

Self-organization of discrete fates in human gastruloids is mediated by a hierarchy of signaling pathways. How these pathways are integrated in time, and whether cells maintain a memory of their signaling history remains obscure. Here, we dissect the temporal integration of two key pathways, WNT and ACTIVIN, which along with BMP control gastrulation. CRISPR/Cas9-engineered live reporters of SMAD1, 2 and 4 demonstrate that in contrast to the stable signaling by SMAD1, signaling and transcriptional response by SMAD2 is transient, and while necessary for pluripotency, it is insufficient for differentiation. Pre-exposure to WNT, however, endows cells with the competence to respond to graded levels of ACTIVIN, which induces differentiation without changing SMAD2 dynamics. This cellular memory of WNT signaling is necessary for ACTIVIN morphogen activity. A re-evaluation of the evidence gathered over decades in model systems, re-enforces our conclusions and points to an evolutionarily conserved mechanism.


Subject(s)
Activins/metabolism , Gastrulation , Wnt Signaling Pathway , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Endoderm/cytology , Genes, Reporter , Humans , Mesoderm/cytology , Mice , Nucleotide Motifs/genetics , Pluripotent Stem Cells/metabolism , Rats , Smad Proteins/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism
5.
Cell Stem Cell ; 23(1): 60-73.e6, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29937203

ABSTRACT

Cortical deep projection neurons (DPNs) are implicated in neurodevelopmental disorders. Although recent findings emphasize post-mitotic programs in projection neuron fate selection, the establishment of primate DPN identity during layer formation is not well understood. The subplate lies underneath the developing cortex and is a post-mitotic compartment that is transiently and disproportionately enlarged in primates in the second trimester. The evolutionary significance of subplate expansion, the molecular identity of its neurons, and its contribution to primate corticogenesis remain open questions. By modeling subplate formation with human pluripotent stem cells (hPSCs), we show that all classes of cortical DPNs can be specified from subplate neurons (SPNs). Post-mitotic WNT signaling regulates DPN class selection, and DPNs in the caudal fetal cortex appear to exclusively derive from SPNs. Our findings indicate that SPNs have evolved in primates as an important source of DPNs that contribute to cortical lamination prior to their known role in circuit formation.


Subject(s)
Cell Differentiation , Cell Lineage , Models, Biological , Neurons/cytology , Pluripotent Stem Cells/cytology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pluripotent Stem Cells/metabolism
6.
Development ; 145(2)2018 01 29.
Article in English | MEDLINE | ID: mdl-29378824

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disease caused by expansion of CAG repeats in the Huntingtin gene (HTT). Neither its pathogenic mechanisms nor the normal functions of HTT are well understood. To model HD in humans, we engineered a genetic allelic series of isogenic human embryonic stem cell (hESC) lines with graded increases in CAG repeat length. Neural differentiation of these lines unveiled a novel developmental HD phenotype: the appearance of giant multinucleated telencephalic neurons at an abundance directly proportional to CAG repeat length, generated by a chromosomal instability and failed cytokinesis over multiple rounds of DNA replication. We conclude that disrupted neurogenesis during development is an important, unrecognized aspect of HD pathogenesis. To address the function of normal HTT protein we generated HTT+/- and HTT-/- lines. Surprisingly, the same phenotype emerged in HTT-/- but not HTT+/- lines. We conclude that HD is a developmental disorder characterized by chromosomal instability that impairs neurogenesis, and that HD represents a genetic dominant-negative loss of function, contrary to the prevalent gain-of-toxic-function hypothesis. The consequences of developmental alterations should be considered as a new target for HD therapies.


Subject(s)
Chromosomal Instability , Huntingtin Protein/genetics , Huntington Disease/genetics , Neurogenesis/genetics , Alleles , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Humans , Huntingtin Protein/deficiency , Huntingtin Protein/metabolism , Huntington Disease/etiology , Huntington Disease/pathology , Models, Biological , Phenotype , Spindle Apparatus/pathology , Trinucleotide Repeat Expansion
7.
Cell Stem Cell ; 21(5): 564-565, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29100008

ABSTRACT

Our understanding of early human development is typically based on inference from animal models, which may not fully recapitulate human embryonic features. As proof of concept, Fogarty et al. (2017) used CRISPR/Cas9 to genetically ablate the OCT4 gene in human preimplantation embryos and found key differences from its function in model systems.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , Blastocyst , Clustered Regularly Interspaced Short Palindromic Repeats , Embryo, Mammalian , Humans
8.
Development ; 144(17): 3042-3053, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28760810

ABSTRACT

Paracrine signals maintain developmental states and create cell fate patterns in vivo and influence differentiation outcomes in human embryonic stem cells (hESCs) in vitro Systematic investigation of morphogen signaling is hampered by the difficulty of disentangling endogenous signaling from experimentally applied ligands. Here, we grow hESCs in micropatterned colonies of 1-8 cells ('µColonies') to quantitatively investigate paracrine signaling and the response to external stimuli. We examine BMP4-mediated differentiation in µColonies and standard culture conditions and find that in µColonies, above a threshold concentration, BMP4 gives rise to only a single cell fate, contrary to its role as a morphogen in other developmental systems. Under standard culture conditions BMP4 acts as a morphogen but this requires secondary signals and particular cell densities. We find that a 'community effect' enforces a common fate within µColonies, both in the state of pluripotency and when cells are differentiated, and that this effect allows a more precise response to external signals. Using live cell imaging to correlate signaling histories with cell fates, we demonstrate that interactions between neighbors result in sustained, homogenous signaling necessary for differentiation.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Signal Transduction/drug effects , Animals , Cell Count , Cell Proliferation/drug effects , Cells, Cultured , Clone Cells , Colony-Forming Units Assay , Ectoderm/cytology , Ectoderm/drug effects , Ectoderm/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Mice , Models, Biological , Nodal Protein/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects
9.
J Mol Biol ; 429(18): 2802-2815, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684247

ABSTRACT

The cell cycle has gained attention as a key determinant for cell fate decisions, but the contribution of DNA replication and mitosis in stem cell differentiation has not been extensively studied. To understand if these processes act as "windows of opportunity" for changes in cell identity, we established synchronized cultures of mouse embryonic stem cells as they exit the ground state of pluripotency. We show that initial transcriptional changes in this transition do not require passage through mitosis and that conversion to primed pluripotency is linked to lineage priming in the G1 phase. Importantly, we demonstrate that impairment of DNA replication severely blocks transcriptional switch to primed pluripotency, even in the absence of p53 activity induced by the DNA damage response. Our data suggest an important role for DNA replication during mouse embryonic stem cell differentiation, which could shed light on why pluripotent cells are only receptive to differentiation signals during G1, that is, before the S phase.


Subject(s)
Cell Differentiation , Cell Division , DNA Replication , Mouse Embryonic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Animals , Mice , Transcription, Genetic
10.
Antiviral Res ; 142: 123-135, 2017 06.
Article in English | MEDLINE | ID: mdl-28359840

ABSTRACT

SAMHD1 is a triphosphohydrolase that restricts HIV-1 by limiting the intracellular dNTP pool required for reverse transcription. Although SAMHD1 is expressed and active/unphosphorylated in most cell lines, its restriction activity is thought to be relevant only in non-cycling cells. However, an in depth evaluation of SAMHD1 function and relevance in cycling cells is required. Here, we show that SAMHD1-induced degradation by HIV-2 Vpx affects the dNTP pool and HIV-1 replication capacity in the presence of the 3'-azido-3'-deoxythymidine (AZT) in cycling cells. Similarly, in SAMHD1 knockout cells, HIV-1 showed increased replicative capacity in the presence of nucleoside inhibitors, especially AZT, that was reverted by re-expression of wild type SAMHD1. Sensitivity to non-nucleoside inhibitors (nevirapine and efavirenz) or the integrase inhibitor raltegravir was not affected by SAMHD1. Combination of three mutations (S18A, T21A, T25A) significantly prevented SAMHD1 phosphorylation but did not significantly affect HIV-1 replication in the presence of AZT. Our results demonstrate that SAMHD1 is active in HIV-1 permissive cells, does not modify susceptibility to HIV-1 infection but strongly affects sensitivity to nucleoside inhibitors.


Subject(s)
HIV-1/drug effects , SAM Domain and HD Domain-Containing Protein 1/pharmacology , Virus Replication/drug effects , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Replication/drug effects , Gene Editing , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , HIV Infections/metabolism , HIV-1/pathogenicity , HIV-2/drug effects , Host-Pathogen Interactions , Humans , Phosphorylation , Reverse Transcription/drug effects , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism , Viral Regulatory and Accessory Proteins/drug effects , Zidovudine/pharmacology
11.
Dev Cell ; 39(3): 302-315, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27746044

ABSTRACT

The earliest aspects of human embryogenesis remain mysterious. To model patterning events in the human embryo, we used colonies of human embryonic stem cells (hESCs) grown on micropatterned substrate and differentiated with BMP4. These gastruloids recapitulate the embryonic arrangement of the mammalian germ layers and provide an assay to assess the structural and signaling mechanisms patterning the human gastrula. Structurally, high-density hESCs localize their receptors to transforming growth factor ß at their lateral side in the center of the colony while maintaining apical localization of receptors at the edge. This relocalization insulates cells at the center from apically applied ligands while maintaining response to basally presented ones. In addition, BMP4 directly induces the expression of its own inhibitor, NOGGIN, generating a reaction-diffusion mechanism that underlies patterning. We develop a quantitative model that integrates edge sensing and inhibitors to predict human fate positioning in gastruloids and, potentially, the human embryo.


Subject(s)
Gastrula/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Animals , Body Patterning/drug effects , Bone Morphogenetic Protein 4/pharmacology , Carrier Proteins/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Polarity/drug effects , Cell Proliferation/drug effects , Colony-Forming Units Assay , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Feedback, Physiological/drug effects , Human Embryonic Stem Cells/drug effects , Humans , Ligands , Mice , Models, Biological , Phosphorylation/drug effects , Receptors, Cell Surface/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad1 Protein/metabolism , Time Factors , Transforming Growth Factor beta/metabolism
12.
Nat Protoc ; 11(11): 2223-2232, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27735934

ABSTRACT

Fate allocation in the gastrulating embryo is spatially organized as cells differentiate into specialized cell types depending on their positions with respect to the body axes. There is a need for in vitro protocols that allow the study of spatial organization associated with this developmental transition. Although embryoid bodies and organoids can exhibit some spatial organization of differentiated cells, methods that generate embryoid bodies or organoids do not yield consistent and fully reproducible results. Here, we describe a micropatterning approach in which human embryonic stem cells are confined to disk-shaped, submillimeter colonies. After 42 h of BMP4 stimulation, cells form self-organized differentiation patterns in concentric radial domains, which express specific markers associated with the embryonic germ layers, reminiscent of gastrulating embryos. Our protocol takes 3 d; it uses commercial microfabricated slides (from CYTOO), human laminin-521 (LN-521) as extracellular matrix coating, and either conditioned or chemically defined medium (mTeSR). Differentiation patterns within individual colonies can be determined by immunofluorescence and analyzed with cellular resolution. Both the size of the micropattern and the type of medium affect the patterning outcome. The protocol is appropriate for personnel with basic stem cell culture training. This protocol describes a robust platform for quantitative analysis of the mechanisms associated with pattern formation at the onset of gastrulation.


Subject(s)
Human Embryonic Stem Cells/cytology , Microtechnology/methods , Cell Differentiation , Cell Line , Gastrulation , Humans
13.
Nat Biotechnol ; 33(12): 1287-1292, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26524661

ABSTRACT

There are numerous cell types with scarcely understood functions, whose interactions with the immune system are not well characterized. To facilitate their study, we generated a mouse bearing enhanced green fluorescent protein (EGFP)-specific CD8+ T cells. Transfer of the T cells into EGFP reporter animals can be used to kill EGFP-expressing cells, allowing selective depletion of desired cell types, or to interrogate T-cell interactions with specific populations. Using this system, we eliminate a rare EGFP-expressing cell type in the heart and demonstrate its role in cardiac function. We also show that naive T cells are recruited into the mouse brain by antigen-expressing microglia, providing evidence of an immune surveillance pathway in the central nervous system. The just EGFP death-inducing (Jedi) T cells enable visualization of a T-cell antigen. They also make it possible to utilize hundreds of existing EGFP-expressing mice, tumors, pathogens and other tools, to study T-cell interactions with many different cell types, to model disease states and to determine the functions of poorly characterized cell populations.

14.
PLoS One ; 10(5): e0127687, 2015.
Article in English | MEDLINE | ID: mdl-26010866

ABSTRACT

Huntington's disease (HD) is a devastating neurological disorder that is caused by an expansion of the poly-Q tract in exon 1 of the Huntingtin gene (HTT). HTT is an evolutionarily conserved and ubiquitously expressed protein that has been linked to a variety of functions including transcriptional regulation, mitochondrial function, and vesicle transport. This large protein has numerous caspase and calpain cleavage sites and can be decorated with several post-translational modifications such as phosphorylations, acetylations, sumoylations, and palmitoylations. However, the exact function of HTT and the role played by its modifications in the cell are still not well understood. Scrutiny of HTT function has been focused on a single, full length mRNA. In this study, we report the discovery of 5 novel HTT mRNA splice isoforms that are expressed in normal and HTT-expanded human embryonic stem cell (hESC) lines as well as in cortical neurons differentiated from hESCs. Interestingly, none of the novel isoforms generates a truncated protein. Instead, 4 of the 5 new isoforms specifically eliminate domains and modifications to generate smaller HTT proteins. The fifth novel isoform incorporates a previously unreported additional exon, dubbed 41b, which is hominid-specific and introduces a potential phosphorylation site in the protein. The discovery of this hominid-specific isoform may shed light on human-specific pathogenic mechanisms of HTT, which could not be investigated with current mouse models of the disease.


Subject(s)
Exons , Huntington Disease , Nerve Tissue Proteins , Animals , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Species Specificity
16.
J Exp Med ; 211(4): 669-83, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24638167

ABSTRACT

Langerhans cell histiocytosis (LCH) is a clonal disorder with elusive etiology, characterized by the accumulation of CD207(+) dendritic cells (DCs) in inflammatory lesions. Recurrent BRAF-V600E mutations have been reported in LCH. In this study, lesions from 100 patients were genotyped, and 64% carried the BRAF-V600E mutation within infiltrating CD207(+) DCs. BRAF-V600E expression in tissue DCs did not define specific clinical risk groups but was associated with increased risk of recurrence. Strikingly, we found that patients with active, high-risk LCH also carried BRAF-V600E in circulating CD11c(+) and CD14(+) fractions and in bone marrow (BM) CD34(+) hematopoietic cell progenitors, whereas the mutation was restricted to lesional CD207(+) DC in low-risk LCH patients. Importantly, BRAF-V600E expression in DCs was sufficient to drive LCH-like disease in mice. Consistent with our findings in humans, expression of BRAF-V600E in BM DC progenitors recapitulated many features of the human high-risk LCH, whereas BRAF-V600E expression in differentiated DCs more closely resembled low-risk LCH. We therefore propose classification of LCH as a myeloid neoplasia and hypothesize that high-risk LCH arises from somatic mutation of a hematopoietic progenitor, whereas low-risk disease arises from somatic mutation of tissue-restricted precursor DCs.


Subject(s)
Cell Differentiation , Dendritic Cells/metabolism , Genetic Predisposition to Disease , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Animals , Antigens, CD34/metabolism , Antigens, Surface/metabolism , Bone Marrow/pathology , CD11c Antigen/metabolism , Cell Lineage , Child , Child, Preschool , Female , Hematopoietic Stem Cells/metabolism , Histiocytosis, Langerhans-Cell/blood , Histocompatibility Antigens Class II/metabolism , Humans , Infant , Lectins, C-Type/metabolism , Male , Mannose-Binding Lectins/metabolism , Mice , Phenotype , Risk Factors , Treatment Outcome
17.
Nat Immunol ; 15(1): 54-62, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24270517

ABSTRACT

miR-126 is a microRNA expressed predominately by endothelial cells and controls angiogenesis. We found miR-126 was required for the innate response to pathogen-associated nucleic acids and that miR-126-deficient mice had greater susceptibility to infection with pseudotyped HIV. Profiling of miRNA indicated that miR-126 had high and specific expression by plasmacytoid dendritic cells (pDCs). Moreover, miR-126 controlled the survival and function of pDCs and regulated the expression of genes encoding molecules involved in the innate response, including Tlr7, Tlr9 and Nfkb1, as well as Kdr, which encodes the growth factor receptor VEGFR2. Deletion of Kdr in DCs resulted in reduced production of type I interferon, which supports the proposal of a role for VEGFR2 in miR-126 regulation of pDCs. Our studies identify the miR-126-VEGFR2 axis as an important regulator of the innate response that operates through multiscale control of pDCs.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , MicroRNAs/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Dendritic Cells/metabolism , Flow Cytometry , HIV Infections/immunology , HIV Infections/virology , Humans , Immunity, Innate/genetics , Immunoblotting , Interferon-alpha/blood , Interferon-alpha/immunology , Interferon-alpha/metabolism , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/immunology , NF-kappa B p50 Subunit/metabolism , Nucleic Acids/immunology , Nucleic Acids/metabolism , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/immunology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Transcriptome/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
J Clin Invest ; 123(8): 3254-3271, 2013 08 01.
Article in English | MEDLINE | ID: mdl-23863627

ABSTRACT

For most lysosomal storage diseases (LSDs) affecting the CNS, there is currently no cure. The BBB, which limits the bioavailability of drugs administered systemically, and the short half-life of lysosomal enzymes, hamper the development of effective therapies. Mucopolysaccharidosis type IIIA (MPS IIIA) is an autosomic recessive LSD caused by a deficiency in sulfamidase, a sulfatase involved in the stepwise degradation of glycosaminoglycan (GAG) heparan sulfate. Here, we demonstrate that intracerebrospinal fluid (intra-CSF) administration of serotype 9 adenoassociated viral vectors (AAV9s) encoding sulfamidase corrects both CNS and somatic pathology in MPS IIIA mice. Following vector administration, enzymatic activity increased throughout the brain and in serum, leading to whole body correction of GAG accumulation and lysosomal pathology, normalization of behavioral deficits, and prolonged survival. To test this strategy in a larger animal, we treated beagle dogs using intracisternal or intracerebroventricular delivery. Administration of sulfamidase-encoding AAV9 resulted in transgenic expression throughout the CNS and liver and increased sulfamidase activity in CSF. High-titer serum antibodies against AAV9 only partially blocked CSF-mediated gene transfer to the brains of dogs. Consistently, anti-AAV antibody titers were lower in CSF than in serum collected from healthy and MPS IIIA-affected children. These results support the clinical translation of this approach for the treatment of MPS IIIA and other LSDs with CNS involvement.

19.
Diabetes ; 62(2): 551-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23099863

ABSTRACT

In type 1 diabetes, loss of tolerance to ß-cell antigens results in T-cell-dependent autoimmune destruction of ß cells. The abrogation of autoreactive T-cell responses is a prerequisite to achieve long-lasting correction of the disease. The liver has unique immunomodulatory properties and hepatic gene transfer results in tolerance induction and suppression of autoimmune diseases, in part by regulatory T-cell (Treg) activation. Hence, the liver could be manipulated to treat or prevent diabetes onset through expression of key genes. IGF-I may be an immunomodulatory candidate because it prevents autoimmune diabetes when expressed in ß cells or subcutaneously injected. Here, we demonstrate that transient, plasmid-derived IGF-I expression in mouse liver suppressed autoimmune diabetes progression. Suppression was associated with decreased islet inflammation and ß-cell apoptosis, increased ß-cell replication, and normalized ß-cell mass. Permanent protection depended on exogenous IGF-I expression in liver nonparenchymal cells and was associated with increased percentage of intrapancreatic Tregs. Importantly, Treg depletion completely abolished IGF-I-mediated protection confirming the therapeutic potential of these cells in autoimmune diabetes. This study demonstrates that a nonviral gene therapy combining the immunological properties of the liver and IGF-I could be beneficial in the treatment of the disease.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Genetic Therapy , Insulin-Like Growth Factor I/genetics , Liver/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Division/genetics , Cell Division/immunology , Humans , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Liver/immunology , Mice , Mice, Transgenic , Pancreatitis/genetics , Pancreatitis/immunology , Plasmids/genetics
20.
Diabetes ; 61(11): 2851-61, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22961079

ABSTRACT

Type 2 diabetes (T2D) results from insulin resistance and inadequate insulin secretion. Insulin resistance initially causes compensatory islet hyperplasia that progresses to islet disorganization and altered vascularization, inflammation, and, finally, decreased functional ß-cell mass and hyperglycemia. The precise mechanism(s) underlying ß-cell failure remain to be elucidated. In this study, we show that in insulin-resistant high-fat diet-fed mice, the enhanced islet vascularization and inflammation was parallel to an increased expression of vascular endothelial growth factor A (VEGF). To elucidate the role of VEGF in these processes, we have genetically engineered ß-cells to overexpress VEGF (in transgenic mice or after adeno-associated viral vector-mediated gene transfer). We found that sustained increases in ß-cell VEGF levels led to disorganized, hypervascularized, and fibrotic islets, progressive macrophage infiltration, and proinflammatory cytokine production, including tumor necrosis factor-α and interleukin-1ß. This resulted in impaired insulin secretion, decreased ß-cell mass, and hyperglycemia with age. These results indicate that sustained VEGF upregulation may participate in the initiation of a process leading to ß-cell failure and further suggest that compensatory islet hyperplasia and hypervascularization may contribute to progressive inflammation and ß-cell mass loss during T2D.


Subject(s)
Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Neovascularization, Pathologic/metabolism , Prediabetic State/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Cytokines/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Disease Progression , Fibrosis , Gene Transfer Techniques , Hyperplasia , Insulin Resistance , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Prediabetic State/etiology , Prediabetic State/immunology , Prediabetic State/pathology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL