Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 11(475)2019 01 16.
Article in English | MEDLINE | ID: mdl-30651324

ABSTRACT

The intestine is a major source of systemic ammonia (NH3); thus, capturing part of gut NH3 may mitigate disease symptoms in conditions of hyperammonemia such as urea cycle disorders and hepatic encephalopathy. As an approach to the lowering of blood ammonia arising from the intestine, we engineered the orally delivered probiotic Escherichia coli Nissle 1917 to create strain SYNB1020 that converts NH3 to l-arginine (l-arg). We up-regulated arginine biosynthesis in SYNB1020 by deleting a negative regulator of l-arg biosynthesis and inserting a feedback-resistant l-arg biosynthetic enzyme. SYNB1020 produced l-arg and consumed NH3 in an in vitro system. SYNB1020 reduced systemic hyperammonemia, improved survival in ornithine transcarbamylase-deficient spfash mice, and decreased hyperammonemia in the thioacetamide-induced liver injury mouse model. A phase 1 clinical study was conducted including 52 male and female healthy adult volunteers. SYNB1020 was well tolerated at daily doses of up to 1.5 × 1012 colony-forming units administered for up to 14 days. A statistically significant dose-dependent increase in urinary nitrate, plasma 15N-nitrate (highest dose versus placebo, P = 0.0015), and urinary 15N-nitrate was demonstrated, indicating in vivo SYNB1020 activity. SYNB1020 concentrations reached steady state by the second day of dosing, and excreted cells were alive and metabolically active as evidenced by fecal arginine production in response to added ammonium chloride. SYNB1020 was no longer detectable in feces 2 weeks after the last dose. These results support further clinical development of SYNB1020 for hyperammonemia disorders including urea cycle disorders and hepatic encephalopathy.


Subject(s)
Escherichia coli/genetics , Genetic Engineering , Healthy Volunteers , Hyperammonemia/therapy , Ammonia/blood , Ammonia/metabolism , Animals , Arginine/metabolism , Biosynthetic Pathways , Disease Models, Animal , Feces/chemistry , Female , Humans , Hyperammonemia/blood , Hyperammonemia/urine , Macaca fascicularis , Male , Mice , Nitrates/blood , Nitrates/urine , Stress, Physiological/genetics , Survival Analysis
2.
Sci Rep ; 8(1): 2554, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416060

ABSTRACT

Ammonia physiology is important to numerous disease states including urea cycle disorders and hepatic encephalopathy. However, many unknowns persist regarding the ammonia response to common and potentially significant physiologic influences, such as food. Our aim was to evaluate the dynamic range of ammonia in response to an oral protein challenge in healthy participants. We measured blood and breath ammonia at baseline and every hour for 5.5 hours. Healthy men (N = 22, aged 18 to 24 years) consumed a 60 g protein shake (high dose); a subset of 10 consumed a 30 g protein shake (moderate dose) and 12 consumed an electrolyte drink containing 0 g protein (control). Change in blood ammonia over time varied by dose (p = 0.001). Difference in blood ammonia was significant for control versus high (p = 0.0004) and moderate versus high (p = 0.03). Change in breath ammonia over time varied by dose (p < 0.0001). Difference in breath ammonia was significant for control versus moderate (p = 0.03) and control versus high (p = 0.0003). Changes in blood and breath ammonia were detectable by fast, minimally-invasive (blood) or non-invasive (breath) point-of-care ammonia measurement methods. These pilot data may contribute to understanding normal ammonia metabolism. Novel measurement methods may aid research into genetic and metabolic ammonia disorders.


Subject(s)
Ammonia/metabolism , Diet, High-Protein , Exhalation , Adolescent , Adult , Ammonia/analysis , Ammonia/blood , Breath Tests , Healthy Volunteers , Humans , Male , Pilot Projects
3.
Clin Transl Sci ; 11(2): 200-207, 2018 03.
Article in English | MEDLINE | ID: mdl-29194983

ABSTRACT

Understanding the pharmacology of microbiome-based therapeutics is required to support the development of new medicines. Strains of E. coli Nissle (EcN) were genetically modified and administered to cynomolgus monkeys at doses of 1 × 109 and 1 × 1012 colony-forming units (CFU)/day for 28 days. A clinical study to evaluate the exposure and clearance of EcN in healthy volunteers was also performed. Healthy subjects received oral doses of EcN, 2.5 to 25 × 109 CFU 3 times daily for 28 days or a single day. In cynomolgus monkeys, replicating strains yielded higher fecal concentrations than nonreplicating strains and persisted for longer following cessation of dosing. In the clinical study, all subjects cleared EcN following cessation of dosing with median clearance of 1 week. Quantitative methodology can be applied to microbiome-based therapeutics, and similar kinetics and clearance were observed for EcN in cynomolgus monkeys and humans.


Subject(s)
Biological Therapy/methods , Escherichia coli/metabolism , Microbiota/physiology , Microorganisms, Genetically-Modified/metabolism , Probiotics/pharmacology , Administration, Oral , Adult , Animals , Arginine/metabolism , Escherichia coli/genetics , Escherichia coli/isolation & purification , Feces/microbiology , Female , Healthy Volunteers , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Male , Metabolic Networks and Pathways/genetics , Metabolism, Inborn Errors/therapy , Microbiota/genetics , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/isolation & purification , Middle Aged , Models, Animal , Primates , Prospective Studies , Young Adult
4.
Nat Biotechnol ; 35(7): 653-658, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553941

ABSTRACT

Bacteria can be engineered to function as diagnostics or therapeutics in the mammalian gut but commercial translation of technologies to accomplish this has been hindered by the susceptibility of synthetic genetic circuits to mutation and unpredictable function during extended gut colonization. Here, we report stable, engineered bacterial strains that maintain their function for 6 months in the mouse gut. We engineered a commensal murine Escherichia coli strain to detect tetrathionate, which is produced during inflammation. Using our engineered diagnostic strain, which retains memory of exposure in the gut for analysis by fecal testing, we detected tetrathionate in both infection-induced and genetic mouse models of inflammation over 6 months. The synthetic genetic circuits in the engineered strain were genetically stable and functioned as intended over time. The durable performance of these strains confirms the potential of engineered bacteria as living diagnostics.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Gastroenteritis/diagnosis , Gastroenteritis/microbiology , Gastrointestinal Microbiome , Tetrathionic Acid/metabolism , Animals , Cell Survival , Escherichia coli/isolation & purification , Female , Genetic Engineering/methods , Intestines , Mice , Mice, Inbred C57BL
5.
Nat Commun ; 6: 10039, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26615910

ABSTRACT

Microbial population growth is typically measured when cells can be directly observed, or when death is rare. However, neither of these conditions hold for the mammalian gut microbiota, and, therefore, standard approaches cannot accurately measure the growth dynamics of this community. Here we introduce a new method (distributed cell division counting, DCDC) that uses the accurate segregation at cell division of genetically encoded fluorescent particles to measure microbial growth rates. Using DCDC, we can measure the growth rate of Escherichia coli for >10 consecutive generations. We demonstrate experimentally and theoretically that DCDC is robust to error across a wide range of temperatures and conditions, including in the mammalian gut. Furthermore, our experimental observations inform a mathematical model of the population dynamics of the gut microbiota. DCDC can enable the study of microbial growth during infection, gut dysbiosis, antibiotic therapy or other situations relevant to human health.


Subject(s)
Cell Division , Cytological Techniques/methods , Escherichia coli/cytology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Escherichia coli/chemistry , Escherichia coli/growth & development , Humans , Kinetics
6.
PLoS One ; 9(4): e93441, 2014.
Article in English | MEDLINE | ID: mdl-24736311

ABSTRACT

UNLABELLED: ß-arrestins, ubiquitous cellular scaffolding proteins that act as signaling mediators of numerous critical cellular pathways, are attractive therapeutic targets because they promote tumorigenesis in several tumor models. However, targeting scaffolding proteins with traditional small molecule drugs has been challenging. Inhibition of ß-arrestin 2 with a novel aptamer impedes multiple oncogenic signaling pathways simultaneously. Additionally, delivery of the ß-arrestin 2-targeting aptamer into leukemia cells through coupling to a recently described cancer cell-specific delivery aptamer, inhibits multiple ß-arrestin-mediated signaling pathways known to be required for chronic myelogenous leukemia (CML) disease progression, and impairs tumorigenic growth in CML patient samples. The ability to target scaffolding proteins such as ß-arrestin 2 with RNA aptamers may prove beneficial as a therapeutic strategy. HIGHLIGHTS: An RNA aptamer inhibits ß-arrestin 2 activity.Inhibiting ß-arrestin 2 impedes multiple tumorigenic pathways simultaneously.The therapeutic aptamer is delivered to cancer cells using a cell-specific DNA aptamer.Targeting ß-arrestin 2 inhibits tumor progression in CML models and patient samples.


Subject(s)
Aptamers, Nucleotide/genetics , Arrestins/genetics , Arrestins/metabolism , Leukemia/genetics , Leukemia/metabolism , Signal Transduction , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , K562 Cells , beta-Arrestin 2 , beta-Arrestins
7.
Proc Natl Acad Sci U S A ; 111(13): 4838-43, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24639514

ABSTRACT

The mammalian gut is a dynamic community of symbiotic microbes that interact with the host to impact health, disease, and metabolism. We constructed engineered bacteria that survive in the mammalian gut and sense, remember, and report on their experiences. Based on previous genetic memory systems, we constructed a two-part system with a "trigger element" in which the lambda Cro gene is transcribed from a tetracycline-inducible promoter, and a "memory element" derived from the cI/Cro region of phage lambda. The memory element has an extremely stable cI state and a Cro state that is stable for many cell divisions. When Escherichia coli bearing the memory system are administered to mice treated with anhydrotetracycline, the recovered bacteria all have switched to the Cro state, whereas those administered to untreated mice remain in the cI state. The trigger and memory elements were transferred from E. coli K12 to a newly isolated murine E. coli strain; the stability and switching properties of the memory element were essentially identical in vitro and during passage through mice, but the engineered murine E. coli was more stably established in the mouse gut. This work lays a foundation for the use of synthetic genetic circuits as monitoring systems in complex, ill-defined environments, and may lead to the development of living diagnostics and therapeutics.


Subject(s)
Escherichia coli/genetics , Gastrointestinal Tract/microbiology , Genetic Engineering , Mammals/microbiology , Microbiota , Animals , Gastrointestinal Tract/drug effects , Humans , Mice , Microbiota/drug effects , Tetracyclines/pharmacology
8.
Nucleic Acid Ther ; 22(3): 187-95, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22703281

ABSTRACT

To reduce the adverse effects of cancer therapies and increase their efficacy, new delivery agents that specifically target cancer cells are needed. We and others have shown that aptamers can selectively deliver therapeutic oligonucleotides to the endosome and cytoplasm of cancer cells that express a particular cell surface receptor. Identifying a single aptamer that can internalize into many different cancer cell-types would increase the utility of aptamer-mediated delivery of therapeutic agents. We investigated the ability of the nucleolin aptamer (AS1411) to internalize into multiple cancer cell types and observed that it internalizes into a wide variety of cancer cells and migrates to the nucleus. To determine if the aptamer could be utilized to deliver therapeutic oligonucleotides to modulate events in the nucleus, we evaluated the ability of the aptamer to deliver splice-switching oligonucleotides. We observed that aptamer-splice-switching oligonucleotide chimeras can alter splicing in the nuclei of treated cells and are effective at lower doses than the splice switching oligonucleotides alone. Our results suggest that aptamers can be utilized to deliver oligonucleotides to the nucleus of a wide variety of cancer cells to modulate nuclear events such as RNA splicing.


Subject(s)
Aptamers, Nucleotide , Cell Nucleus/metabolism , Oligonucleotides/administration & dosage , RNA Splicing , Cell Line, Tumor , Endocytosis , ErbB Receptors/genetics , Flow Cytometry , Humans , Neoplasms/metabolism , Oligonucleotides/metabolism , Phosphoproteins/genetics , RNA Interference , RNA-Binding Proteins/genetics , Nucleolin
SELECTION OF CITATIONS
SEARCH DETAIL
...