Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Diabetologia ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38871836

ABSTRACT

AIMS/HYPOTHESIS: Stem cell-derived islets (SC-islets) are being used as cell replacement therapy for insulin-dependent diabetes. Non-invasive long-term monitoring methods for SC-islet grafts, which are needed to detect misguided differentiation in vivo and to optimise their therapeutic effectiveness, are lacking. Positron emission tomography (PET) has been used to monitor transplanted primary islets. We therefore aimed to apply PET as a non-invasive monitoring method for SC-islet grafts. METHODS: We implanted different doses of human SC-islets, SC-islets derived using an older protocol or a state-of-the-art protocol and SC-islets genetically rendered hyper- or hypoactive into mouse calf muscle to yield different kinds of grafts. We followed the grafts with PET using two tracers, glucagon-like peptide 1 receptor-binding [18F]F-dibenzocyclooctyne-exendin-4 ([18F]exendin) and the dopamine precursor 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine ([18F]FDOPA), for 5 months, followed by histological assessment of graft size and composition. Additionally, we implanted a kidney subcapsular cohort with different SC-islet doses to assess the connection between C-peptide and stem cell-derived beta cell (SC-beta cell) mass. RESULTS: Small but pure and large but impure grafts were derived from SC-islets. PET imaging allowed detection of SC-islet grafts even <1 mm3 in size, [18F]exendin having a better detection rate than [18F]FDOPA (69% vs 44%, <1 mm3; 96% vs 85%, >1 mm3). Graft volume quantified with [18F]exendin (r2=0.91) and [18F]FDOPA (r2=0.86) strongly correlated with actual graft volume. [18F]exendin PET delineated large cystic structures and its uptake correlated with graft SC-beta cell proportion (r2=0.68). The performance of neither tracer was affected by SC-islet graft hyper- or hypoactivity. C-peptide measurements under fasted or glucose-stimulated conditions did not correlate with SC-islet graft volume or SC-beta cell mass, with C-peptide under hypoglycaemia having a weak correlation with SC-beta cell mass (r2=0.52). CONCLUSIONS/INTERPRETATION: [18F]exendin and [18F]FDOPA PET enable non-invasive assessment of SC-islet graft size and aspects of graft composition. These methods could be leveraged for optimising SC-islet cell replacement therapy in diabetes.

2.
Diabetologia ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743124

ABSTRACT

AIMS/HYPOTHESIS: Regulatory factor X 6 (RFX6) is crucial for pancreatic endocrine development and differentiation. The RFX6 variant p.His293LeufsTer7 is significantly enriched in the Finnish population, with almost 1:250 individuals as a carrier. Importantly, the FinnGen study indicates a high predisposition for heterozygous carriers to develop type 2 and gestational diabetes. However, the precise mechanism of this predisposition remains unknown. METHODS: To understand the role of this variant in beta cell development and function, we used CRISPR technology to generate allelic series of pluripotent stem cells. We created two isogenic stem cell models: a human embryonic stem cell model; and a patient-derived stem cell model. Both were differentiated into pancreatic islet lineages (stem-cell-derived islets, SC-islets), followed by implantation in immunocompromised NOD-SCID-Gamma mice. RESULTS: Stem cell models of the homozygous variant RFX6-/- predictably failed to generate insulin-secreting pancreatic beta cells, mirroring the phenotype observed in Mitchell-Riley syndrome. Notably, at the pancreatic endocrine stage, there was an upregulation of precursor markers NEUROG3 and SOX9, accompanied by increased apoptosis. Intriguingly, heterozygous RFX6+/- SC-islets exhibited RFX6 haploinsufficiency (54.2% reduction in protein expression), associated with reduced beta cell maturation markers, altered calcium signalling and impaired insulin secretion (62% and 54% reduction in basal and high glucose conditions, respectively). However, RFX6 haploinsufficiency did not have an impact on beta cell number or insulin content. The reduced insulin secretion persisted after in vivo implantation in mice, aligning with the increased risk of variant carriers to develop diabetes. CONCLUSIONS/INTERPRETATION: Our allelic series isogenic SC-islet models represent a powerful tool to elucidate specific aetiologies of diabetes in humans, enabling the sensitive detection of aberrations in both beta cell development and function. We highlight the critical role of RFX6 in augmenting and maintaining the pancreatic progenitor pool, with an endocrine roadblock and increased cell death upon its loss. We demonstrate that RFX6 haploinsufficiency does not affect beta cell number or insulin content but does impair function, predisposing heterozygous carriers of loss-of-function variants to diabetes. DATA AVAILABILITY: Ultra-deep bulk RNA-seq data for pancreatic differentiation stages 3, 5 and 7 of H1 RFX6 genotypes are deposited in the Gene Expression Omnibus database with accession code GSE234289. Original western blot images are deposited at Mendeley ( https://data.mendeley.com/datasets/g75drr3mgw/2 ).

3.
Diabetes ; 73(7): 1127-1139, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38603470

ABSTRACT

Pluripotent stem cell-derived islets (SC-islets) have emerged as a new source for ß-cell replacement therapy. The function of human islet transplants is hampered by excessive cell death posttransplantation; contributing factors include inflammatory reactions, insufficient revascularization, and islet amyloid formation. However, there is a gap in knowledge of the engraftment process of SC-islets. In this experimental study, we investigated the engraftment capability of SC-islets at 3 months posttransplantation and observed that cell apoptosis rates were lower but vascular density was similar in SC-islets compared with human islets. Whereas the human islet transplant vascular structures were a mixture of remnant donor endothelium and ingrowing blood vessels, the SC-islets contained ingrowing blood vessels only. Oxygenation in the SC-islet grafts was twice as high as that in the corresponding grafts of human islets, suggesting better vascular functionality. Similar to the blood vessel ingrowth, reinnervation of the SC-islets was four- to fivefold higher than that of the human islets. Both SC-islets and human islets contained amyloid at 1 and 3 months posttransplantation. We conclude that the vascular and neural engraftment of SC-islets are superior to those of human islets, but grafts of both origins develop amyloid, with potential long-term consequences.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Islets of Langerhans Transplantation/methods , Islets of Langerhans/blood supply , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Animals , Mice , Apoptosis/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Graft Survival/physiology , Male
4.
Nat Genet ; 55(12): 2075-2081, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37973953

ABSTRACT

Identifying genes linked to extreme phenotypes in humans has the potential to highlight biological processes not shared with all other mammals. Here, we report the identification of homozygous loss-of-function variants in the primate-specific gene ZNF808 as a cause of pancreatic agenesis. ZNF808 is a member of the KRAB zinc finger protein family, a large and rapidly evolving group of epigenetic silencers which target transposable elements. We show that loss of ZNF808 in vitro results in aberrant activation of regulatory potential contained in the primate-specific transposable elements it represses during early pancreas development. This leads to inappropriate specification of cell fate with induction of genes associated with liver identity. Our results highlight the essential role of ZNF808 in pancreatic development in humans and the contribution of primate-specific regions of the human genome to congenital developmental disease.


Subject(s)
Congenital Abnormalities , DNA Transposable Elements , DNA-Binding Proteins , Pancreas , Animals , Humans , Cell Differentiation , Genome, Human , Primates/abnormalities , Primates/genetics , DNA-Binding Proteins/genetics , Congenital Abnormalities/genetics , Pancreas/abnormalities
5.
Nat Cell Biol ; 24(10): 1528-1540, 2022 10.
Article in English | MEDLINE | ID: mdl-36202974

ABSTRACT

The biological purpose of long non-coding RNAs (lncRNAs) is poorly understood. Haploinsufficient mutations in HNF1A homeobox A (HNF1A), encoding a homeodomain transcription factor, cause diabetes mellitus. Here, we examine HASTER, the promoter of an lncRNA antisense to HNF1A. Using mouse and human models, we show that HASTER maintains cell-specific physiological HNF1A concentrations through positive and negative feedback loops. Pancreatic ß cells from Haster mutant mice consequently showed variegated HNF1A silencing or overexpression, resulting in hyperglycaemia. HASTER-dependent negative feedback was essential to prevent HNF1A binding to inappropriate genomic regions. We demonstrate that the HASTER promoter DNA, rather than the lncRNA, modulates HNF1A promoter-enhancer interactions in cis and thereby regulates HNF1A transcription. Our studies expose a cis-regulatory element that is unlike classic enhancers or silencers, it stabilizes the transcription of its target gene and ensures the fidelity of a cell-specific transcription factor program. They also show that disruption of a mammalian lncRNA promoter can cause diabetes mellitus.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha , Promoter Regions, Genetic , RNA, Long Noncoding , Animals , Humans , Mice , Hepatocyte Nuclear Factor 1-alpha/genetics , Mammals , RNA, Long Noncoding/genetics , Transcription, Genetic/genetics , Transcription, Genetic/physiology
6.
Nat Commun ; 13(1): 6363, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289205

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that results in the destruction of insulin producing pancreatic ß-cells. One of the genes associated with T1D is TYK2, which encodes a Janus kinase with critical roles in type-Ι interferon (IFN-Ι) mediated intracellular signalling. To study the role of TYK2 in ß-cell development and response to IFNα, we generated TYK2 knockout human iPSCs and directed them into the pancreatic endocrine lineage. Here we show that loss of TYK2 compromises the emergence of endocrine precursors by regulating KRAS expression, while mature stem cell-islets (SC-islets) function is not affected. In the SC-islets, the loss or inhibition of TYK2 prevents IFNα-induced antigen processing and presentation, including MHC Class Ι and Class ΙΙ expression, enhancing their survival against CD8+ T-cell cytotoxicity. These results identify an unsuspected role for TYK2 in ß-cell development and support TYK2 inhibition in adult ß-cells as a potent therapeutic target to halt T1D progression.


Subject(s)
Diabetes Mellitus, Type 1 , Insulins , Humans , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Insulins/metabolism , Interferon-alpha/pharmacology , Interferon-alpha/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Insulin-Secreting Cells
7.
Differentiation ; 128: 83-100, 2022.
Article in English | MEDLINE | ID: mdl-36114074

ABSTRACT

Nuclear receptor subfamily 5 group A member 1 (NR5A1) encodes steroidogenic factor 1 (SF1), a key regulatory factor that determines gonadal development and coordinates endocrine functions. Here, we have established a stem cell-based model of human gonadal development and applied it to evaluate the effects of NR5A1 during the transition from bipotential gonad to testicular cells. We combined directed differentiation of human induced pluripotent stem cells (46,XY) with activation of endogenous NR5A1 expression by conditionally-inducible CRISPR activation. The resulting male gonadal-like cells expressed several Sertoli cell transcripts, secreted anti-Müllerian hormone and responded to follicle-stimulating hormone by producing sex steroid intermediates. These characteristics were not induced without NR5A1 activation. A total of 2691 differentially expressed genetic elements, including both coding and non-coding RNAs, were detected immediately following activation of NR5A1 expression. Of those, we identified novel gonad-related putative NR5A1 targets, such as SCARA5, which we validated also by immunocytochemistry. In addition, NR5A1 activation was associated with dynamic expression of multiple gonad- and infertility-related differentially expressed genes. In conclusion, by combining targeted differentiation and endogenous activation of NR5A1 we have for the first time, been able to examine in detail the effects of NR5A1 in early human gonadal cells. The model and results obtained provide a useful resource for future investigations exploring the causative reasons for gonadal dysgenesis and infertility in humans.


Subject(s)
Induced Pluripotent Stem Cells , Infertility , Humans , Male , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Mutation , Induced Pluripotent Stem Cells/metabolism , Gonads/metabolism , Scavenger Receptors, Class A/genetics
8.
STAR Protoc ; 3(4): 101711, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36136756

ABSTRACT

We present here a robust and reliable protocol by which to differentiate pancreatic islet-like aggregates (SC-islets) from human pluripotent stem cells. The 7-stage protocol mimics developmental patterning factors that induce endocrine lineage formation and spans monolayer, microwell, and aggregate suspension culture. The SC-islets demonstrate dynamic glucose-sensitive insulin secretion and an endocrine cell composition similar to those of primary human islets. SC-islets generated using this optimized protocol are suitable for in vitro modeling of islet cell pathophysiology and therapeutic applications. For complete details on the use and execution of this protocol, please refer to Balboa et al. (2022).


Subject(s)
Islets of Langerhans , Pluripotent Stem Cells , Humans , Cell Differentiation/physiology , Glucose/metabolism , Insulin Secretion
9.
Dev Cell ; 57(16): 1922-1936.e9, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35998583

ABSTRACT

Sequence variants in cis-acting enhancers are important for polygenic disease, but their role in Mendelian disease is poorly understood. Redundancy between enhancers that regulate the same gene is thought to mitigate the pathogenic impact of enhancer mutations. Recent findings, however, have shown that loss-of-function mutations in a single enhancer near PTF1A cause pancreas agenesis and neonatal diabetes. Using mouse and human genetic models, we show that this enhancer activates an entire PTF1A enhancer cluster in early pancreatic multipotent progenitors. This leading role, therefore, precludes functional redundancy. We further demonstrate that transient expression of PTF1A in multipotent progenitors sets in motion an epigenetic cascade that is required for duct and endocrine differentiation. These findings shed insights into the genome regulatory mechanisms that drive pancreas differentiation. Furthermore, they reveal an enhancer that acts as a regulatory master key and is thus vulnerable to pathogenic loss-of-function mutations.


Subject(s)
Diabetes Mellitus , Transcription Factors , Animals , Cell Differentiation/genetics , Diabetes Mellitus/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Humans , Infant, Newborn , Mice , Mutation/genetics , Pancreas/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
10.
Nat Biotechnol ; 40(7): 1042-1055, 2022 07.
Article in English | MEDLINE | ID: mdl-35241836

ABSTRACT

Transplantation of pancreatic islet cells derived from human pluripotent stem cells is a promising treatment for diabetes. Despite progress in the generation of stem-cell-derived islets (SC-islets), no detailed characterization of their functional properties has been conducted. Here, we generated functionally mature SC-islets using an optimized protocol and benchmarked them comprehensively against primary adult islets. Biphasic glucose-stimulated insulin secretion developed during in vitro maturation, associated with cytoarchitectural reorganization and the increasing presence of alpha cells. Electrophysiology, signaling and exocytosis of SC-islets were similar to those of adult islets. Glucose-responsive insulin secretion was achieved despite differences in glycolytic and mitochondrial glucose metabolism. Single-cell transcriptomics of SC-islets in vitro and throughout 6 months of engraftment in mice revealed a continuous maturation trajectory culminating in a transcriptional landscape closely resembling that of primary islets. Our thorough evaluation of SC-islet maturation highlights their advanced degree of functionality and supports their use in further efforts to understand and combat diabetes.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans , Pluripotent Stem Cells , Animals , Glucose/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Mice , Pluripotent Stem Cells/metabolism
11.
Stem Cell Reports ; 17(2): 413-426, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35063129

ABSTRACT

Conventional reprogramming methods rely on the ectopic expression of transcription factors to reprogram somatic cells into induced pluripotent stem cells (iPSCs). The forced expression of transcription factors may lead to off-target gene activation and heterogeneous reprogramming, resulting in the emergence of alternative cell types and aberrant iPSCs. Activation of endogenous pluripotency factors by CRISPR activation (CRISPRa) can reduce this heterogeneity. Here, we describe a high-efficiency reprogramming of human somatic cells into iPSCs using optimized CRISPRa. Efficient reprogramming was dependent on the additional targeting of the embryo genome activation-enriched Alu-motif and the miR-302/367 locus. Single-cell transcriptome analysis revealed that the optimized CRISPRa reprogrammed cells more directly and specifically into the pluripotent state when compared to the conventional reprogramming method. These findings support the use of CRISPRa for high-quality pluripotent reprogramming of human cells.


Subject(s)
Cellular Reprogramming/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Alu Elements/genetics , Gene Expression Profiling , Genetic Loci , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , Single-Cell Analysis
12.
Front Endocrinol (Lausanne) ; 12: 642152, 2021.
Article in English | MEDLINE | ID: mdl-33828531

ABSTRACT

Diabetes mellitus is characterized by elevated levels of blood glucose and is ultimately caused by insufficient insulin production from pancreatic beta cells. Different research models have been utilized to unravel the molecular mechanisms leading to the onset of diabetes. The generation of pancreatic endocrine cells from human pluripotent stem cells constitutes an approach to study genetic defects leading to impaired beta cell development and function. Here, we review the recent progress in generating and characterizing functional stem cell-derived beta cells. We summarize the diabetes disease modeling possibilities that stem cells offer and the challenges that lie ahead to further improve these models.


Subject(s)
Diabetes Mellitus/metabolism , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Pluripotent Stem Cells/cytology , Animals , CRISPR-Cas Systems , Cell Culture Techniques , Cell Differentiation , Diabetes Mellitus/physiopathology , Disease Models, Animal , Endocrine Cells/metabolism , Genetic Variation , Genomics , Glucose/metabolism , Humans , Induced Pluripotent Stem Cells , Insulin/metabolism , Islets of Langerhans/metabolism , Phosphorylation , Proteomics , Transcriptome
13.
Diabetes ; 70(4): 1006-1018, 2021 04.
Article in English | MEDLINE | ID: mdl-33500254

ABSTRACT

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-resident protein that plays a crucial role in attenuating ER stress responses. Although MANF is indispensable for the survival and function of mouse ß-cells, its precise role in human ß-cell development and function is unknown. In this study, we show that lack of MANF in humans results in diabetes due to increased ER stress, leading to impaired ß-cell function. We identified two patients from different families with childhood diabetes and a neurodevelopmental disorder associated with homozygous loss-of-function mutations in the MANF gene. To study the role of MANF in human ß-cell development and function, we knocked out the MANF gene in human embryonic stem cells and differentiated them into pancreatic endocrine cells. Loss of MANF induced mild ER stress and impaired insulin-processing capacity of ß-cells in vitro. Upon implantation to immunocompromised mice, the MANF knockout grafts presented elevated ER stress and functional failure, particularly in recipients with diabetes. By describing a new form of monogenic neurodevelopmental diabetes syndrome caused by disturbed ER function, we highlight the importance of adequate ER stress regulation for proper human ß-cell function and demonstrate the crucial role of MANF in this process.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Nerve Growth Factors/metabolism , Blotting, Western , Endoplasmic Reticulum Stress/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Editing/methods , Glucose Tolerance Test , Humans , Immunohistochemistry , Male , Mutation/genetics , Nerve Growth Factors/genetics , Real-Time Polymerase Chain Reaction , Streptozocin/pharmacology
14.
Diabetologia ; 64(3): 630-640, 2021 03.
Article in English | MEDLINE | ID: mdl-33404684

ABSTRACT

AIMS/HYPOTHESIS: Congenital hyperinsulinism caused by mutations in the KATP-channel-encoding genes (KATPHI) is a potentially life-threatening disorder of the pancreatic beta cells. No optimal medical treatment is available for patients with diazoxide-unresponsive diffuse KATPHI. Therefore, we aimed to create a model of KATPHI using patient induced pluripotent stem cell (iPSC)-derived islets. METHODS: We derived iPSCs from a patient carrying a homozygous ABCC8V187D mutation, which inactivates the sulfonylurea receptor 1 (SUR1) subunit of the KATP-channel. CRISPR-Cas9 mutation-corrected iPSCs were used as controls. Both were differentiated to stem cell-derived islet-like clusters (SC-islets) and implanted into NOD-SCID gamma mice. RESULTS: SUR1-mutant and -corrected iPSC lines both differentiated towards the endocrine lineage, but SUR1-mutant stem cells generated 32% more beta-like cells (SC-beta cells) (64.6% vs 49.0%, p = 0.02) and 26% fewer alpha-like cells (16.1% vs 21.8% p = 0.01). SUR1-mutant SC-beta cells were 61% more proliferative (1.23% vs 0.76%, p = 0.006), and this phenotype could be induced in SUR1-corrected cells with pharmacological KATP-channel inactivation. The SUR1-mutant SC-islets secreted 3.2-fold more insulin in low glucose conditions (0.0174% vs 0.0054%/min, p = 0.0021) and did not respond to KATP-channel-acting drugs in vitro. Mice carrying grafts of SUR1-mutant SC-islets presented with 38% lower fasting blood glucose (4.8 vs 7.7 mmol/l, p = 0.009) and their grafts failed to efficiently shut down insulin secretion during induced hypoglycaemia. Explanted SUR1-mutant grafts displayed an increase in SC-beta cell proportion and SC-beta cell nucleomegaly, which was independent of proliferation. CONCLUSIONS/INTERPRETATION: We have created a model recapitulating the known pathophysiology of KATPHI both in vitro and in vivo. We have also identified a novel role for KATP-channel activity during human islet development. This model will enable further studies for the improved understanding and clinical management of KATPHI without the need for primary patient tissue.


Subject(s)
Congenital Hyperinsulinism/metabolism , Induced Pluripotent Stem Cells/metabolism , Islets of Langerhans/metabolism , Sulfonylurea Receptors/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/pathology , Congenital Hyperinsulinism/physiopathology , Female , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/transplantation , Insulin Secretion , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Islets of Langerhans Transplantation , Male , Mice, Inbred NOD , Mice, SCID , Mutation , Phenotype , Sulfonylurea Receptors/genetics
15.
Viruses ; 12(9)2020 08 27.
Article in English | MEDLINE | ID: mdl-32867368

ABSTRACT

CRISPR activation (CRISPRa) has revealed great potential as a tool to modulate the expression of targeted cellular genes. Here, we successfully applied the CRISPRa system to trigger the Kaposi's sarcoma-associated herpesvirus (KSHV) reactivation in latently infected cells by selectively activating ORF50 gene directly from the virus genome. We found that a nuclease-deficient Cas9 (dCas9) fused to a destabilization domain (DD) and 12 copies of the VP16 activation domain (VP192) triggered a more efficient KSHV lytic cycle and virus production when guided to two different sites on the ORF50 promoter, instead of only a single site. To our surprise, the virus reactivation induced by binding of the stable DD-dCas9-VP192 on the ORF50 promoter was even more efficient than reactivation induced by ectopic expression of ORF50. This suggests that recruitment of additional transcriptional activators to the ORF50 promoter, in addition to ORF50 itself, are needed for the efficient virus production. Further, we show that CRISPRa can be applied to selectively express the early lytic gene, ORF57, without disturbing the viral latency. Therefore, CRISPRa-based systems can be utilized to facilitate virus-host interaction studies by controlling the expression of not only cellular but also of specific KSHV genes.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Herpesvirus 8, Human/physiology , Immediate-Early Proteins/genetics , Sarcoma, Kaposi/virology , Trans-Activators/genetics , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/metabolism , Promoter Regions, Genetic , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Virus Activation , Virus Latency
16.
Dis Model Mech ; 13(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-31996360

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons provide a fundamental signal for the onset of puberty and subsequent reproductive functions by secretion of gonadotropin-releasing hormone. Their disrupted development or function leads to congenital hypogonadotropic hypogonadism (CHH). To model the development of human GnRH neurons, we generated a stable GNRH1-TdTomato reporter cell line in human pluripotent stem cells (hPSCs) using CRISPR-Cas9 genome editing. RNA-sequencing of the reporter clone, differentiated into GnRH neurons by dual SMAD inhibition and FGF8 treatment, revealed 6461 differentially expressed genes between progenitors and GnRH neurons. Expression of the transcription factor ISL1, one of the top 50 most upregulated genes in the TdTomato-expressing GnRH neurons, was confirmed in 10.5 gestational week-old human fetal GnRH neurons. Among the differentially expressed genes, we detected 15 genes that are implicated in CHH and several genes that are implicated in human puberty timing. Finally, FGF8 treatment in the neuronal progenitor pool led to upregulation of 37 genes expressed both in progenitors and in TdTomato-expressing GnRH neurons, which suggests upstream regulation of these genes by FGF8 signaling during GnRH neuron differentiation. These results illustrate how hPSC-derived human GnRH neuron transcriptomic analysis can be utilized to dissect signaling pathways and gene regulatory networks involved in human GnRH neuron development.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Genes, Reporter , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Transcriptome/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cell Line , Fetus/cytology , Fibroblast Growth Factor 8/pharmacology , Humans , Hypogonadism/genetics , LIM-Homeodomain Proteins/metabolism , Neurons/drug effects , Pluripotent Stem Cells/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
17.
F1000Res ; 9: 1336, 2020.
Article in English | MEDLINE | ID: mdl-34745570

ABSTRACT

The COVID-19 pandemic has posed and is continuously posing enormous societal and health challenges worldwide. The research community has mobilized to develop novel projects to find a cure or a vaccine, as well as to contribute to mass testing, which has been a critical measure to contain the infection in several countries. Through this article, we share our experiences and learnings as a group of volunteers at the Centre for Genomic Regulation (CRG) in Barcelona, Spain. As members of the ORFEU project, an initiative by the Government of Catalonia to achieve mass testing of people at risk and contain the epidemic in Spain, we share our motivations, challenges and the key lessons learnt, which we feel will help better prepare the global society to address similar situations in the future.


Subject(s)
COVID-19 , COVID-19 Testing , Genomics , Humans , Pandemics , SARS-CoV-2 , Volunteers
18.
Diabetologia ; 62(8): 1329-1336, 2019 08.
Article in English | MEDLINE | ID: mdl-31161346

ABSTRACT

Understanding the molecular mechanisms behind beta cell dysfunction is essential for the development of effective and specific approaches for diabetes care and prevention. Physiological human beta cell models are needed for this work. We review the possibilities and limitations of currently available human beta cell models and how they can be dramatically enhanced using genome-editing technologies. In addition to the gold standard, primary isolated islets, other models now include immortalised human beta cell lines and pluripotent stem cell-derived islet-like cells. The scarcity of human primary islet samples limits their use, but valuable gene expression and functional data from large collections of human islets have been made available to the scientific community. The possibilities for studying beta cell physiology using immortalised human beta cell lines and stem cell-derived islets are rapidly evolving. However, the functional immaturity of these cells is still a significant limitation. CRISPR-Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9) has enabled precise engineering of specific genetic variants, targeted transcriptional modulation and genome-wide genetic screening. These approaches can now be exploited to gain understanding of the mechanisms behind coding and non-coding diabetes-associated genetic variants, allowing more precise evaluation of their contribution to diabetes pathogenesis. Despite all the progress, genome editing in primary pancreatic islets remains difficult to achieve, an important limitation requiring further technological development.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/therapy , Gene Editing , Genome, Human , Insulin-Secreting Cells/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Gene Editing/trends , Gene Silencing , Genetic Variation , Genotype , Humans , Pluripotent Stem Cells , Polymorphism, Single Nucleotide , Risk
20.
Nat Commun ; 10(1): 620, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30728358

ABSTRACT

Elevated MYC expression sensitizes tumor cells to apoptosis but the therapeutic potential of this mechanism remains unclear. We find, in a model of MYC-driven breast cancer, that pharmacological activation of AMPK strongly synergizes with BCL-2/BCL-XL inhibitors to activate apoptosis. We demonstrate the translational potential of an AMPK and BCL-2/BCL-XL co-targeting strategy in ex vivo and in vivo models of MYC-high breast cancer. Metformin combined with navitoclax or venetoclax efficiently inhibited tumor growth, conferred survival benefits and induced tumor infiltration by immune cells. However, withdrawal of the drugs allowed tumor re-growth with presentation of PD-1+/CD8+ T cell infiltrates, suggesting immune escape. A two-step treatment regimen, beginning with neoadjuvant metformin+venetoclax to induce apoptosis and followed by adjuvant metformin+venetoclax+anti-PD-1 treatment to overcome immune escape, led to durable antitumor responses even after drug withdrawal. We demonstrate that pharmacological reactivation of MYC-dependent apoptosis is a powerful antitumor strategy involving both tumor cell depletion and immunosurveillance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Genes, myc/drug effects , Immunotherapy , Aniline Compounds/pharmacology , Animals , Antibodies, Monoclonal, Humanized , Apoptosis/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CD8-Positive T-Lymphocytes , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Combinations , Female , HEK293 Cells , Heterografts , Humans , Metformin/pharmacology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2 , Sulfonamides/pharmacology , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...