Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Ophthalmic Res ; 66(1): 878-884, 2023.
Article in English | MEDLINE | ID: mdl-37094557

ABSTRACT

INTRODUCTION: Retinitis pigmentosa (RP) is a rare degenerative retinal disease caused by mutations in approximately seventy genes. Currently, despite the availability of large-scale DNA sequencing technologies, ∼30-40% of patients still cannot be diagnosed at the molecular level. In this study, we investigated a novel intronic deletion of PDE6B, encoding the beta subunit of phosphodiesterase 6 in association with recessive RP. METHODS: Three unrelated consanguineous families were recruited from the northwestern part of Pakistan. Whole exome sequencing was performed for the proband of each family, and the data were analyzed according to an in-house computer pipeline. Relevant DNA variants in all available members of these families were assessed through Sanger sequencing. A minigene-based splicing assay was also performed. RESULTS: The clinical phenotype for all patients was compatible with rod cone degeneration, with the onset during childhood. Whole exome sequencing revealed a homozygous 18 bp intronic deletion (NM_000283.3:c.1921-20_1921-3del) in PDE6B, which co-segregated with disease in 10 affected individuals. In vitro splicing tests showed that this deletion causes aberrant RNA splicing of the gene, leading to the in-frame deletion of 6 codons and, likely, to disease. CONCLUSION: Our findings further expand the mutational spectrum of the PDE6B gene.


Subject(s)
Retinitis Pigmentosa , Humans , DNA Mutational Analysis , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Mutation , RNA Splicing , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Pedigree , Eye Proteins/genetics
2.
J Control Release ; 352: 994-1008, 2022 12.
Article in English | MEDLINE | ID: mdl-36370877

ABSTRACT

Wireless powered optogenetic cell-based implant provides a strategy to deliver subcutaneously therapeutic proteins. Immortalize Human Mesenchymal Stem Cells (hMSC-TERT) expressing the bacteriophytochrome diguanylate cyclase (DGCL) were validated for optogenetic controlled interferon-ß delivery (Optoferon cells) in a bioelectronic cell-based implant. Optoferon cells transcriptomic profiling was used to elaborate an in-silico model of the recombinant interferon-ß production. Wireless optoelectronic device integration was developed using additive manufacturing and injection molding. Implant cell-based optoelectronic interface manufacturing was established to integrate industrial flexible compact low-resistance screen-printed Near Field Communication (NFC) coil antenna. Optogenetic cell-based implant biocompatibility, and device performances were evaluated in the Experimental Autoimmune Encephalomyelitis (EAE) mouse model of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Humans , Multiple Sclerosis/therapy , Encephalomyelitis, Autoimmune, Experimental/therapy , Interferon-beta/genetics , Interferon-beta/metabolism , Disease Models, Animal , Gene Expression , Mice, Inbred C57BL
3.
Hum Mol Genet ; 31(12): 1970-1978, 2022 06 22.
Article in English | MEDLINE | ID: mdl-34999892

ABSTRACT

In the framework of the UK 100 000 Genomes Project, we investigated the genetic origin of a previously undescribed recessive dermatological condition, which we named LIPHAK (LTV1-associated Inflammatory Poikiloderma with Hair abnormalities and Acral Keratoses), in four affected individuals from two UK families of Pakistani and Indian origins, respectively. Our analysis showed that only one gene, LTV1, carried rare biallelic variants that were shared in all affected individuals, and specifically they bore the NM_032860.5:c.503A > G, p.(Asn168Ser) change, found homozygously in all of them. In addition, high-resolution homozygosity mapping revealed the presence of a small 652-kb stretch on chromosome 6, encompassing LTV1, that was haploidentical and common to all affected individuals. The c.503A > G variant was predicted by in silico tools to affect the correct splicing of LTV1's exon 5. Minigene-driven splicing assays in HEK293T cells and in a skin sample from one of the patients confirmed that this variant was indeed responsible for the creation of a new donor splice site, resulting in aberrant splicing and in a premature termination codon in exon 6 of this gene. LTV1 encodes one of the ribosome biogenesis factors that promote the assembly of the small (40S) ribosomal subunit. In yeast, defects in LTV1 alter the export of nascent ribosomal subunits to the cytoplasm; however, the role of this gene in human pathology is unknown to date. Our data suggest that LIPHAK could be a previously unrecognized ribosomopathy.


Subject(s)
Hair Diseases , Ribosomes , Skin Diseases , Humans , Hair Diseases/genetics , HEK293 Cells , Mutation , Ribosomes/genetics , Skin Diseases/genetics , Syndrome
4.
Cornea ; 41(1): 89-94, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34469340

ABSTRACT

PURPOSE: The purpose of this study was to investigate the clinical and genetic features of a man and his daughter with posterior polymorphous corneal dystrophy (PPCD), referred to our clinic for Descemet membrane endothelial keratoplasty. No other known relatives were affected. METHODS: Ophthalmic examination and histology, including electron microscopy, were performed. Genetic testing was conducted by means of whole exome sequencing, and variant analysis was achieved by using an internal in silico pipeline. Molecular tests included a dual-luciferase assay. RESULTS: Slowly progressive blurred vision was reported from childhood by the daughter. The father's symptoms started at age 55. Best-corrected visual acuity was reduced in both patients (0.2-0.4). Slit-lamp examination in both patients revealed bilateral corneal clouding with gray endothelial lesions; other family members had no ophthalmological signs. Descemet membrane endothelial keratoplasty was performed uneventfully in both patients. Histology showed thickened Descemet membrane and abnormal endothelium resembling epithelial-like cells. Both patients carried the OVOL2 5' untranslated region NM_021220.4.c.-61G>A variant in the heterozygous state. This change was associated with increased promoter activity and was not present in the unaffected members of the family. CONCLUSIONS: The 5' untranslated region mutation c.-61G>A in OVOL2 has been previously found in 1 individual with PPCD1 and reported as a variant of unknown significance because of insufficient evidence supporting its pathogenicity. Identification of the second family with 2 individuals affected by PPCD1 carrying this change, together with functional data, provides further proofs that it is disease-causing.


Subject(s)
5' Untranslated Regions/genetics , Corneal Dystrophies, Hereditary/genetics , Endothelium, Corneal/ultrastructure , Mutation , Transcription Factors/genetics , Adult , Aged , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/metabolism , DNA Mutational Analysis , Endothelium, Corneal/pathology , Female , Humans , Male , Microscopy, Electron , Pedigree , Promoter Regions, Genetic , Slit Lamp Microscopy , Zinc Fingers
5.
Pharmacoeconomics ; 40(1): 91-108, 2022 01.
Article in English | MEDLINE | ID: mdl-34480325

ABSTRACT

BACKGROUND: Current first-line disease-modifying therapies (DMT) for multiple sclerosis (MS) patients are injectable or oral treatments. The Optogenerapy consortium is developing a novel bioelectronic cell-based implant for controlled release of beta-interferon (IFNß1a) protein into the body. The current study estimated the potential cost effectiveness of the Optogenerapy implant (hereafter: Optoferon) compared with injectable IFNß1a (Avonex). METHODS: A Markov model simulating the costs and effects of Optoferon compared with injectable 30 mg IFNß1a over a 9-year time horizon from a Dutch societal perspective. Costs were reported in 2019 Euros and discounted at a 4% annual rate; health effects were discounted at a 1.5% annual rate. The cohort consisted of 35-year-old, relapsing-remitting MS patients with mild disability. The device is implanted in a daycare setting, and is replaced every 3 years. In the base-case analysis, we assumed equal input parameters for Optoferon and Avonex regarding disability progression, health effects, adverse event probabilities, and acquisition costs. We assumed reduced annual relapse rates and withdrawal rates for Optoferon compared with Avonex. Sensitivity, scenario, value of information, and headroom analysis were performed. RESULTS: Optoferon was the dominant strategy with cost reductions (- €26,966) and health gains (0.45 quality-adjusted life-years gained). A main driver of cost differences are the acquisition costs of Optoferon being 2.5 times less than the costs of Avonex. The incremental cost-effectiveness ratio was most sensitive to variations in the annual acquisition costs of Avonex, the annual withdrawal rate of Avonex and Optoferon, and the disability progression of Avonex. CONCLUSION: Innovative technology such as the Optoferon implant may be a cost-effective therapy for patients with MS. The novel implantable mode of therapeutic protein administration has the potential to become a new mode of treatment administration for MS patients and in other disease areas. However, trials are needed to establish safety and effectiveness.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Adult , Cost-Benefit Analysis , Humans , Interferon beta-1a , Interferon-beta , Multiple Sclerosis/drug therapy , Multiple Sclerosis, Relapsing-Remitting/drug therapy
6.
Front Bioeng Biotechnol ; 9: 620967, 2021.
Article in English | MEDLINE | ID: mdl-34249877

ABSTRACT

Improving a drug delivery system is critical to treat central nervous system disorders. Here we studied an innovative approach based on implantation of a wireless-powered cell-based device in mice. This device, coupling biologic material and electronics, is the first of its kind. The advantage of this technology is its ability to control the secretion of a therapeutic molecule and to switch the classical permanent delivery to activation on demand. In diseases with relapsing-remitting phases such as multiple sclerosis, such activation could be selectively achieved in relapsing phases. However, the safety (tolerance to biomaterials and surgical procedure) of such a clinical device needs to be verified. Therefore, the development of tools to assess the biocompatibility of the system in animal models is an essential step. We present the development of this new therapeutic approach, the challenges we encountered during the different steps of its development (such as cell loading in the chamber, surgery protocol for subcutaneous implantation of the device) and the tools we used to evaluate cell viability and biocompatibility of the device.

7.
PLoS Genet ; 16(12): e1009201, 2020 12.
Article in English | MEDLINE | ID: mdl-33383577

ABSTRACT

Conjunctival melanoma (CJM) is a rare but potentially lethal and highly-recurrent cancer of the eye. Similar to cutaneous melanoma (CM), it originates from melanocytes. Unlike CM, however, CJM is relatively poorly characterized from a genomic point of view. To fill this knowledge gap and gain insight into the genomic nature of CJM, we performed whole-exome (WES) or whole-genome sequencing (WGS) of tumor-normal tissue pairs in 14 affected individuals, as well as RNA sequencing in a subset of 11 tumor tissues. Our results show that, similarly to CM, CJM is also characterized by a very high mutation load, composed of approximately 500 somatic mutations in exonic regions. This, as well as the presence of a UV light-induced mutational signature, are clear signs of the role of sunlight in CJM tumorigenesis. In addition, the genomic classification of CM proposed by TCGA seems to be well-applicable to CJM, with the presence of four typical subclasses defined on the basis of the most frequently mutated genes: BRAF, NF1, RAS, and triple wild-type. In line with these results, transcriptomic analyses revealed similarities with CM as well, namely the presence of a transcriptomic subtype enriched for immune genes and a subtype enriched for genes associated with keratins and epithelial functions. Finally, in seven tumors we detected somatic mutations in ACSS3, a possible new candidate oncogene. Transfected conjunctival melanoma cells overexpressing mutant ACSS3 showed higher proliferative activity, supporting the direct involvement of this gene in the tumorigenesis of CJM. Altogether, our results provide the first unbiased and complete genomic and transcriptomic classification of CJM.


Subject(s)
Conjunctival Neoplasms/genetics , DNA Copy Number Variations , Melanoma/genetics , Mutation , Transcriptome , Cell Line, Tumor , Conjunctival Neoplasms/metabolism , Female , Humans , Male , Melanoma/metabolism , Middle Aged , Neurofibromin 1/genetics , Proto-Oncogene Proteins B-raf/genetics , ras Proteins/genetics
8.
Sci Eng Ethics ; 25(1): 53, 2019 02.
Article in English | MEDLINE | ID: mdl-29368114

ABSTRACT

The author group of above-mentioned review paper was incorrectly published in the online article.

9.
Sci Eng Ethics ; 25(1): 33-52, 2019 02.
Article in English | MEDLINE | ID: mdl-29255953

ABSTRACT

Advances at the interface between the biological sciences and engineering are giving rise to emerging research fields such as synthetic biology. Harnessing the potential of synthetic biology requires timely and adequate translation into clinical practice. However, the translational research enterprise is currently facing fundamental obstacles that slow down the transition of scientific discoveries from the laboratory to the patient bedside. These obstacles including scarce financial resources and deficiency of organizational and logistic settings are widely discussed as primary impediments to translational research. In addition, a number of socio-ethical considerations inherent in translational research need to be addressed. As the translational capacity of synthetic biology is tightly linked to its social acceptance and ethical approval, ethical limitations may-together with financial and organizational problems-be co-determinants of suboptimal translation. Therefore, an early assessment of such limitations will contribute to proactively favor successful translation and prevent the promising potential of synthetic biology from remaining under-expressed. Through the discussion of two case-specific inventions in synthetic biology and their associated ethical implications, we illustrate the socio-ethical challenges ahead in the process of implementing synthetic biology into clinical practice. Since reducing the translational lag is essential for delivering the benefits of basic biomedical research to society at large and promoting global health, we advocate a moral obligation to accelerating translational research: the "translational imperative."


Subject(s)
Inventions/ethics , Synthetic Biology/ethics , Translational Research, Biomedical/ethics , Bioethics , Humans
10.
Cell ; 174(2): 259-270.e11, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29937224

ABSTRACT

Many community- and hospital-acquired bacterial infections are caused by antibiotic-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) predisposes humans to invasive infections that are difficult to eradicate. We designed a closed-loop gene network programming mammalian cells to autonomously detect and eliminate bacterial infections. The genetic circuit contains human Toll-like receptors as the bacterial sensor and a synthetic promoter driving reversible and adjustable expression of lysostaphin, a bacteriolytic enzyme highly lethal to S. aureus. Immunomimetic designer cells harboring this genetic circuit exhibited fast and robust sense-and-destroy kinetics against live staphylococci. When tested in a foreign-body infection model in mice, microencapsulated cell implants prevented planktonic MRSA infection and reduced MRSA biofilm formation by 91%. Notably, this system achieved a 100% cure rate of acute MRSA infections, whereas conventional vancomycin treatment failed. These results suggest that immunomimetic designer cells could offer a therapeutic approach for early detection, prevention, and cure of pathogenic infections in the post-antibiotic era.


Subject(s)
Biomimetics/methods , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/prevention & control , Alkaline Phosphatase/blood , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Disk Diffusion Antimicrobial Tests , Female , HEK293 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lysostaphin/metabolism , Lysostaphin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Transcription Factor AP-1/metabolism
11.
Sci Transl Med ; 10(437)2018 04 18.
Article in English | MEDLINE | ID: mdl-29669854

ABSTRACT

Diagnosis marks the beginning of any successful therapy. Because many medical conditions progress asymptomatically over extended periods of time, their timely diagnosis remains difficult, and this adversely affects patient prognosis. Focusing on hypercalcemia associated with cancer, we aimed to develop a synthetic biology-inspired biomedical tattoo using engineered cells that would (i) monitor long-term blood calcium concentration, (ii) detect onset of mild hypercalcemia, and (iii) respond via subcutaneous accumulation of the black pigment melanin to form a visible tattoo. For this purpose, we designed cells containing an ectopically expressed calcium-sensing receptor rewired to a synthetic signaling cascade that activates expression of transgenic tyrosinase, which produces melanin in response to persistently increased blood Ca2+ We confirmed that the melanin-generated color change produced by this biomedical tattoo could be detected with the naked eye and optically quantified. The system was validated in wild-type mice bearing subcutaneously implanted encapsulated engineered cells. All animals inoculated with hypercalcemic breast and colon adenocarcinoma cells developed tattoos, whereas no tattoos were seen in animals inoculated with normocalcemic tumor cells. All tumor-bearing animals remained asymptomatic throughout the 38-day experimental period. Although hypercalcemia is also associated with other pathologies, our findings demonstrate that it is possible to detect hypercalcemia associated with cancer in murine models using this cell-based diagnostic strategy.


Subject(s)
Calcium/blood , Hypercalcemia/blood , Hypercalcemia/diagnosis , Synthetic Biology/methods , Animals , Breast Neoplasms/blood , Cell Line , Colonic Neoplasms/blood , Female , Humans , Hypercalcemia/etiology , Melanins/blood , Mice , Neoplasms/blood , Neoplasms/complications
12.
Nat Chem Biol ; 13(3): 309-316, 2017 03.
Article in English | MEDLINE | ID: mdl-28092361

ABSTRACT

Synthetic biology advances the rational engineering of mammalian cells to achieve cell-based therapy goals. Synthetic gene networks have nearly reached the complexity of digital electronic circuits and enable single cells to perform programmable arithmetic calculations or to provide dynamic remote control of transgenes through electromagnetic waves. We designed a synthetic multilayered gaseous-fragrance-programmable analog-to-digital converter (ADC) allowing for remote control of digital gene expression with 2-bit AND-, OR- and NOR-gate logic in synchronized cell consortia. The ADC consists of multiple sampling-and-quantization modules sensing analog gaseous fragrance inputs; a gas-to-liquid transducer converting fragrance intensity into diffusible cell-to-cell signaling compounds; a digitization unit with a genetic amplifier circuit to improve the signal-to-noise ratio; and recombinase-based digital expression switches enabling 2-bit processing of logic gates. Synthetic ADCs that can remotely control cellular activities with digital precision may enable the development of novel biosensors and may provide bioelectronic interfaces synchronizing analog metabolic pathways with digital electronics.


Subject(s)
Analog-Digital Conversion , Odorants/analysis , Synthetic Biology/methods , Cells, Cultured , HEK293 Cells , Humans , Perfume/analysis
13.
Porto Biomed J ; 2(5): 145-149, 2017.
Article in English | MEDLINE | ID: mdl-32258609

ABSTRACT

Resort to medications dates back million years ago with the use of medicinal plants. In the nineteenth century, significant contributions in medicine appeared in different domains, among which the invention of a specific drug delivery device; the syringe. Nowadays, injection therapy of bio-manufactured drugs is routine practice for chronic diseases but remains constraining and painful. New emerging advanced therapies invest in genetic, electronics and cell-based therapy for addressing unmet needs for the caregivers and the patient. As digital process in health (eHealth) gains momentum, connected advanced bio-electronic devices now offer new strategies for personalized injection therapies. In this review, we take a journey along the genesis path of a new drug delivery system: the Optogenerapy, a synergy between optogenetic and gene therapy. Inside a bio-electronic implant, electronics and optogenetics are interfaced by light as a traceless inducer signal. By controlling a synthetic optogenetic pathway in the cell, therapeutics delivery can be fine-tuned with a precise spatiotemporal control. The technology holds promise of a new modern syringe era capable of producing a drug of interest at will directly inside the patient.

14.
Nat Commun ; 7: 11247, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27063289

ABSTRACT

Synthetic biology has advanced the design of standardized transcription control devices that programme cellular behaviour. By coupling synthetic signalling cascade- and transcription factor-based gene switches with reverse and differential sensitivity to the licensed food additive vanillic acid, we designed a synthetic lineage-control network combining vanillic acid-triggered mutually exclusive expression switches for the transcription factors Ngn3 (neurogenin 3; OFF-ON-OFF) and Pdx1 (pancreatic and duodenal homeobox 1; ON-OFF-ON) with the concomitant induction of MafA (V-maf musculoaponeurotic fibrosarcoma oncogene homologue A; OFF-ON). This designer network consisting of different network topologies orchestrating the timely control of transgenic and genomic Ngn3, Pdx1 and MafA variants is able to programme human induced pluripotent stem cells (hIPSCs)-derived pancreatic progenitor cells into glucose-sensitive insulin-secreting beta-like cells, whose glucose-stimulated insulin-release dynamics are comparable to human pancreatic islets. Synthetic lineage-control networks may provide the missing link to genetically programme somatic cells into autologous cell phenotypes for regenerative medicine.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Glucose/pharmacology , Induced Pluripotent Stem Cells/cytology , Insulin-Secreting Cells/cytology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Middle Aged , Reproducibility of Results , Transcription Factors/metabolism , Vanillic Acid/pharmacology
15.
Proc Natl Acad Sci U S A ; 113(5): 1244-9, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787873

ABSTRACT

Graves' disease is an autoimmune disorder that causes hyperthyroidism because of autoantibodies that bind to the thyroid-stimulating hormone receptor (TSHR) on the thyroid gland, triggering thyroid hormone release. The physiological control of thyroid hormone homeostasis by the feedback loops involving the hypothalamus-pituitary-thyroid axis is disrupted by these stimulating autoantibodies. To reset the endogenous thyrotrophic feedback control, we designed a synthetic mammalian gene circuit that maintains thyroid hormone homeostasis by monitoring thyroid hormone levels and coordinating the expression of a thyroid-stimulating hormone receptor antagonist (TSHAntag), which competitively inhibits the binding of thyroid-stimulating hormone or the human autoantibody to TSHR. This synthetic control device consists of a synthetic thyroid-sensing receptor (TSR), a yeast Gal4 protein/human thyroid receptor-α fusion, which reversibly triggers expression of the TSHAntag gene from TSR-dependent promoters. In hyperthyroid mice, this synthetic circuit sensed pathological thyroid hormone levels and restored the thyrotrophic feedback control of the hypothalamus-pituitary-thyroid axis to euthyroid hormone levels. Therapeutic plug and play gene circuits that restore physiological feedback control in metabolic disorders foster advanced gene- and cell-based therapies.


Subject(s)
Disease Models, Animal , Gene Regulatory Networks , Genes, Synthetic , Graves Disease/genetics , Pituitary Gland/physiopathology , Thyroid Gland/physiopathology , Animals , Cells, Cultured , Feedback , Graves Disease/physiopathology , Humans , Mice , Thyroid Hormones/blood
16.
J Biotechnol ; 219: 72-85, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26686315

ABSTRACT

The production of therapeutic antibodies using mammalian cells remains a high-priority in the biopharmaceutical manufacturing industry. Bioengineers have targeted different cellular processes, including transcription, translation, secretion and post-translational modifications, to overcome the metabolic bottlenecks limiting production capacity and create high-producing mammalian cell lines. The polycomb group (PcG) proteins belong to a family of chromatin regulators with important roles in multicellular development. By overexpressing and screening genes from the PcG family, we have identified an epigenetic key player for biopharmaceutical manufacturing enhancement: the transcription factor Yin Yang 1 (YY1). The overexpression of YY1 led to an increase in the production of several product genes (SEAP, VEGF165, IgG including Rituximab), provided that human YY1 (hYY1) was expressed in human cells (HeLa, HT-1080, HEK-293T, FreeStyle™ 293-F) and Chinese hamster ovary cell-derived YY1 (cYY1) was expressed in CHO cells (CHO-K1, CHO-easyC, FreeStyle™ CHO-S, CHO-B13-24, CHO-IgG1). Ectopic expression of cYY1 in the stable CHO-derived IgG producer cell lines CHO-B13-24 and CHO-IgG1 increased the antibody titer up to 6-fold, suggesting that epigenetic engineering of mammalian production cell lines could become a new strategy to improve the manufacturing of complex protein pharmaceuticals.


Subject(s)
Antibodies/metabolism , Gene Expression , Protein Engineering/methods , YY1 Transcription Factor/genetics , Animals , Antibodies/genetics , CHO Cells , Cricetulus , Epigenesis, Genetic , HeLa Cells , Humans
17.
Metab Eng ; 29: 169-179, 2015 May.
Article in English | MEDLINE | ID: mdl-25843350

ABSTRACT

Cyclic guanosine monophosphate (cGMP) is a universal second messenger that is synthesized from guanosine triphosphate (GTP) by guanylyl cyclases (GCs) and hydrolyzed into guanosine monophosphate (GMP) by phosphodiesterases (PDEs). Small-molecule drugs that induce high cGMP levels in specialized tissues by boosting GC activity or inhibiting PDE activity have become the predominant treatment strategy for a wide range of medical conditions, including congestive heart failure, pulmonary hypertension, atherosclerosis-based claudication and erectile dysfunction. By fusing the cGMP receptor protein (CRP) of Rhodospirillum centenum to the Herpes simplex-derived transactivation domain VP16, we created a novel synthetic mammalian cGMP-sensing transcription factor (GTA) that activates synthetic promoters (PGTA) containing newly identified GTA-specific operator sites in a concentration-dependent manner. In cell lines expressing endogenous natriuretic peptide receptor A (NPR-A) (HeLa), GTA/PGTA-driven transgene expression was induced by B-type natriuretic peptide (BNP; Nesiritide(®)) in a concentration-dependent manner, which activated NPR-A׳s intracellular GC domain and triggered a corresponding cGMP surge. Ectopic expression of NPR-A in NPR-A-negative cell lines (HEK-293T) produced high cGMP levels and mediated maximum GTA/PGTA-driven transgene expression, which was suppressed by co-expression of PDEs (PDE-3A, PDE-5A and PDE-9A) and was re-triggered by the corresponding PDE inhibitor drugs (Pletal(®), Perfan(®), Primacor(®) (PDE-3A), Viagra(®), Levitra(®), Cialis(®) (PDE-5A) and BAY73-6691 (PDE-9A)). Mice implanted with microencapsulated designer cells co-expressing the GTA/PGTA device with NPR-A and PDE-5A showed control of blood SEAP levels through administration of sildenafil (Viagra(®)). Designer cells engineered for PDE inhibitor-modulated transgene expression may provide a cell-based PDE-targeting drug discovery platform and enable drug-adjusted gene- and cell-based therapies.


Subject(s)
Bacterial Proteins , Carrier Proteins , Cyclic GMP/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Rhodospirillum centenum/genetics , Sildenafil Citrate/pharmacology , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Mice
18.
Angew Chem Int Ed Engl ; 54(20): 5933-8, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25788334

ABSTRACT

Precise spatiotemporal control of physiological processes by optogenetic devices inspired by synthetic biology may provide novel treatment opportunities for gene- and cell-based therapies. An erectile optogenetic stimulator (EROS), a synthetic designer guanylate cyclase producing a blue-light-inducible surge of the second messenger cyclic guanosine monophosphate (cGMP) in mammalian cells, enabled blue-light-dependent penile erection associated with occasional ejaculation after illumination of EROS-transfected corpus cavernosum in male rats. Photostimulated short-circuiting of complex psychological, neural, vascular, and endocrine factors to stimulate penile erection in the absence of sexual arousal may foster novel advances in the treatment of erectile dysfunction.


Subject(s)
Light , Optogenetics/methods , Penile Erection/drug effects , Animals , Cyclic GMP/biosynthesis , Cyclic GMP/metabolism , Erectile Dysfunction/drug therapy , Guanylate Cyclase/metabolism , Male , Rats
19.
Nat Commun ; 5: 5392, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25386727

ABSTRACT

Synthetic devices for traceless remote control of gene expression may provide new treatment opportunities in future gene- and cell-based therapies. Here we report the design of a synthetic mind-controlled gene switch that enables human brain activities and mental states to wirelessly programme the transgene expression in human cells. An electroencephalography (EEG)-based brain-computer interface (BCI) processing mental state-specific brain waves programs an inductively linked wireless-powered optogenetic implant containing designer cells engineered for near-infrared (NIR) light-adjustable expression of the human glycoprotein SEAP (secreted alkaline phosphatase). The synthetic optogenetic signalling pathway interfacing the BCI with target gene expression consists of an engineered NIR light-activated bacterial diguanylate cyclase (DGCL) producing the orthogonal second messenger cyclic diguanosine monophosphate (c-di-GMP), which triggers the stimulator of interferon genes (STING)-dependent induction of synthetic interferon-ß promoters. Humans generating different mental states (biofeedback control, concentration, meditation) can differentially control SEAP production of the designer cells in culture and of subcutaneous wireless-powered optogenetic implants in mice.


Subject(s)
Brain-Computer Interfaces , Gene Expression , Implants, Experimental , Optogenetics/methods , Transgenes , Alkaline Phosphatase/biosynthesis , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Electroencephalography , Female , Humans , Mice , Signal Transduction , Transcription, Genetic , Wireless Technology
20.
J Biol Chem ; 288(48): 34514-28, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24126912

ABSTRACT

WhiB-like (Wbl) proteins are well known for their diverse roles in actinobacterial morphogenesis, cell division, virulence, primary and secondary metabolism, and intrinsic antibiotic resistance. Gene disruption experiments showed that three different Actinobacteria (Mycobacterium smegmatis, Streptomyces lividans, and Rhodococcus jostii) each exhibited a different whiB7-dependent resistance profile. Heterologous expression of whiB7 genes showed these resistance profiles reflected the host's repertoire of endogenous whiB7-dependent genes. Transcriptional activation of two resistance genes in the whiB7 regulon, tap (a multidrug transporter) and erm(37) (a ribosomal methyltransferase), required interaction of WhiB7 with their promoters. Furthermore, heterologous expression of tap genes isolated from Mycobacterium species demonstrated that divergencies in drug specificity of homologous structural proteins contribute to the variation of WhiB7-dependent drug resistance. WhiB7 has a specific tryptophan/glycine-rich region and four conserved cysteine residues; it also has a peptide sequence (AT-hook) at its C terminus that binds AT-rich DNA sequence motifs upstream of the promoters it activates. Targeted mutagenesis showed that these motifs were required to provide antibiotic resistance in vivo. Anaerobically purified WhiB7 from S. lividans was dimeric and contained 2.1 ± 0.3 and 2.2 ± 0.3 mol of iron and sulfur, respectively, per protomer (consistent with the presence of a 2Fe-2S cluster). However, the properties of the dimer's absorption spectrum were most consistent with the presence of an oxygen-labile 4Fe-4S cluster, suggesting 50% occupancy. These data provide the first insights into WhiB7 iron-sulfur clusters as they exist in vivo, a major unresolved issue in studies of Wbl proteins.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Iron-Sulfur Proteins/genetics , Mycobacterium smegmatis/metabolism , Rhodococcus/metabolism , Streptomyces lividans/metabolism , Transcription Factors/genetics , Actinobacteria , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions/genetics , Iron-Sulfur Proteins/metabolism , Methyltransferases/metabolism , Mutagenesis , Mycobacterium smegmatis/genetics , Rhodococcus/genetics , Species Specificity , Streptomyces lividans/genetics , Transcription Factors/isolation & purification , Transcription Factors/metabolism , Transcriptional Activation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...