Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Mol Syst Biol ; 18(3): e10539, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35253995

RESUMO

Enteric hyperoxaluria (EH) is a metabolic disease caused by excessive absorption of dietary oxalate leading to the formation of chronic kidney stones and kidney failure. There are no approved pharmaceutical treatments for EH. SYNB8802 is an engineered bacterial therapeutic designed to consume oxalate in the gut and lower urinary oxalate as a potential treatment for EH. Oral administration of SYNB8802 leads to significantly decreased urinary oxalate excretion in healthy mice and non-human primates, demonstrating the strain's ability to consume oxalate in vivo. A mathematical modeling framework was constructed that combines in vitro and in vivo preclinical data to predict the effects of SYNB8802 administration on urinary oxalate excretion in humans. Simulations of SYNB8802 administration predict a clinically meaningful lowering of urinary oxalate excretion in healthy volunteers and EH patients. Together, these findings suggest that SYNB8802 is a promising treatment for EH.


Assuntos
Hiperoxalúria , Animais , Simulação por Computador , Feminino , Humanos , Hiperoxalúria/etiologia , Hiperoxalúria/urina , Masculino , Camundongos , Oxalatos/metabolismo , Oxalatos/urina
3.
Commun Biol ; 4(1): 898, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294862

RESUMO

The development of therapeutics depends on predictions of clinical activity from pre-clinical data. We have previously described SYNB1618, an engineered bacterial therapeutic (synthetic biotic) for the treatment of Phenylketonuria (PKU), a rare genetic disease that leads to accumulation of plasma phenylalanine (Phe) and severe neurological complications. SYNB1618 consumes Phe in preclinical models, healthy human volunteers, and PKU patients. However, it remains unclear to what extent Phe consumption by SYNB1618 in the gastrointestinal tract lowers plasma Phe levels in PKU patients. Here, we construct a mechanistic model that predicts SYNB1618 function in non-human primates and healthy subjects by combining in vitro simulations and prior knowledge of human physiology. In addition, we extend a model of plasma Phe kinetics in PKU patients, in order to estimate plasma Phe lowering by SYNB1618. This approach provides a framework that can be used more broadly to define the therapeutic potential of synthetic biotics.


Assuntos
Voluntários Saudáveis , Fenilcetonúrias/genética , Primatas/fisiologia , Animais , Humanos , Fenilcetonúrias/metabolismo , Primatas/genética
4.
Nat Metab ; 3(8): 1125-1132, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34294923

RESUMO

Phenylketonuria (PKU) is a rare disease caused by biallelic mutations in the PAH gene that result in an inability to convert phenylalanine (Phe) to tyrosine, elevated blood Phe levels and severe neurological complications if untreated. Most patients are unable to adhere to the protein-restricted diet, and thus do not achieve target blood Phe levels. We engineered a strain of E. coli Nissle 1917, designated SYNB1618, through insertion of the genes encoding phenylalanine ammonia lyase and L-amino acid deaminase into the genome, which allow for bacterial consumption of Phe within the gastrointestinal tract. SYNB1618 was studied in a phase 1/2a randomized, placebo-controlled, double-blind, multi-centre, in-patient study ( NCT03516487 ) in adult healthy volunteers (n = 56) and patients with PKU and blood Phe level ≥600 mmol l-1 (n = 14). Participants were randomized to receive a single dose of SYNB1618 or placebo (part 1) or up to three times per day for up to 7 days (part 2). The primary outcome of this study was safety and tolerability, and the secondary outcome was microbial kinetics. A D5-Phe tracer (15 mg kg-1) was used to study exploratory pharmacodynamic effects. SYNB1618 was safe and well tolerated with a maximum tolerated dose of 2 × 1011 colony-forming units. Adverse events were mostly gastrointestinal and of mild to moderate severity. All participants cleared the bacteria within 4 days of the last dose. Dose-responsive increases in strain-specific Phe metabolites in plasma (trans-cinnamic acid) and urine (hippuric acid) were observed, providing a proof of mechanism for the potential to use engineered bacteria in the treatment of rare metabolic disorders.


Assuntos
Terapia Biológica/métodos , Escherichia coli , Fenilcetonúrias/terapia , Amidoidrolases/genética , Amidoidrolases/metabolismo , Terapia Biológica/efeitos adversos , Escherichia coli/enzimologia , Escherichia coli/genética , Engenharia Genética , Humanos , Fenilalanina Amônia-Liase/genética , Fenilalanina Amônia-Liase/metabolismo , Fenilcetonúrias/sangue , Fenilcetonúrias/genética , Resultado do Tratamento
5.
Nat Commun ; 12(1): 2805, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990606

RESUMO

Engineered bacteria (synthetic biotics) represent a new class of therapeutics that leverage the tools of synthetic biology. Translational testing strategies are required to predict synthetic biotic function in the human body. Gut-on-a-chip microfluidics technology presents an opportunity to characterize strain function within a simulated human gastrointestinal tract. Here, we apply a human gut-chip model and a synthetic biotic designed for the treatment of phenylketonuria to demonstrate dose-dependent production of a strain-specific biomarker, to describe human tissue responses to the engineered strain, and to show reduced blood phenylalanine accumulation after administration of the engineered strain. Lastly, we show how in vitro gut-chip models can be used to construct mechanistic models of strain activity and recapitulate the behavior of the engineered strain in a non-human primate model. These data demonstrate that gut-chip models, together with mechanistic models, provide a framework to predict the function of candidate strains in vivo.


Assuntos
Bactérias/genética , Bactérias/metabolismo , Terapia Biológica/métodos , Microbioma Gastrointestinal , Dispositivos Lab-On-A-Chip , Modelos Biológicos , Fenilcetonúrias/terapia , Animais , Células CACO-2 , Simulação por Computador , Escherichia coli/metabolismo , Engenharia Genética , Células HT29 , Humanos , Técnicas In Vitro , Microfluídica , Fenilalanina/metabolismo , Fenilcetonúrias/metabolismo , Fenilcetonúrias/microbiologia , Primatas , Biologia Sintética
6.
Nat Commun ; 11(1): 1738, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32269218

RESUMO

A complex interplay of metabolic and immunological mechanisms underlies many diseases that represent a substantial unmet medical need. There is an increasing appreciation of the role microbes play in human health and disease, and evidence is accumulating that a new class of live biotherapeutics comprised of engineered microbes could address specific mechanisms of disease. Using the tools of synthetic biology, nonpathogenic bacteria can be designed to sense and respond to environmental signals in order to consume harmful compounds and deliver therapeutic effectors. In this perspective, we describe considerations for the design and development of engineered live biotherapeutics to achieve regulatory and patient acceptance.


Assuntos
Bactérias/genética , Doença , Engenharia Genética , Biomarcadores/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Neoplasias/terapia
7.
Sci Transl Med ; 11(475)2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651324

RESUMO

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.


Assuntos
Escherichia coli/genética , Engenharia Genética , Voluntários Saudáveis , Hiperamonemia/terapia , Amônia/sangue , Amônia/metabolismo , Animais , Arginina/metabolismo , Vias Biossintéticas , Modelos Animais de Doenças , Fezes/química , Feminino , Humanos , Hiperamonemia/sangue , Hiperamonemia/urina , Macaca fascicularis , Masculino , Camundongos , Nitratos/sangue , Nitratos/urina , Estresse Fisiológico/genética , Análise de Sobrevida
8.
Nature ; 535(7610): 48-55, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27383979

RESUMO

When most people think of human development, they tend to consider only human cells and organs. Yet there is another facet that involves human-associated microbial communities. A microbial perspective of human development provides opportunities to refine our definitions of healthy prenatal and postnatal growth and to develop innovative strategies for disease prevention and treatment. Given the dramatic changes in lifestyles and disease patterns that are occurring with globalization, we issue a call for the establishment of 'human microbial observatories' designed to examine microbial community development in birth cohorts representing populations with diverse anthropological characteristics, including those undergoing rapid change.


Assuntos
Biologia do Desenvolvimento , Microbiota/fisiologia , Feminino , Feto/microbiologia , Microbioma Gastrointestinal , Humanos , Lactente , Leite Humano/química , Leite Humano/microbiologia , Boca/microbiologia , Gravidez , Vagina/microbiologia , Desmame
9.
Science ; 352(6293): 1533, 2016 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-27339978

RESUMO

Childhood undernutrition is a major global health challenge. Although current therapeutic approaches have reduced mortality in individuals with severe disease, they have had limited efficacy in ameliorating long-term sequelae, notably stunting, immune dysfunction, and neurocognitive deficits. Recent work is providing insights about the role of impaired development of the human gut microbiota in disease pathogenesis, leading to new concepts for treatment and prevention. These findings raise intriguing basic questions about the mechanisms that direct normal gut microbial community assembly and functional maturation. Designing and implementing new microbiota-directed therapeutics for undernutrition highlights the need to simultaneously consider a variety of features of human biology as well as broader societal issues.


Assuntos
Microbioma Gastrointestinal/fisiologia , Desnutrição/microbiologia , Desnutrição/terapia , Consórcios Microbianos/fisiologia , Animais , Carga Bacteriana , Criança , Desenvolvimento Infantil , Modelos Animais de Doenças , Exposição Ambiental , Vida Livre de Germes , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Desnutrição/complicações , Camundongos , Leite Humano/química , Oligossacarídeos/análise
10.
Cell ; 164(5): 859-71, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26898329

RESUMO

Identifying interventions that more effectively promote healthy growth of children with undernutrition is a pressing global health goal. Analysis of human milk oligosaccharides (HMOs) from 6-month-postpartum mothers in two Malawian birth cohorts revealed that sialylated HMOs are significantly less abundant in those with severely stunted infants. To explore this association, we colonized young germ-free mice with a consortium of bacterial strains cultured from the fecal microbiota of a 6-month-old stunted Malawian infant and fed recipient animals a prototypic Malawian diet with or without purified sialylated bovine milk oligosaccharides (S-BMO). S-BMO produced a microbiota-dependent augmentation of lean body mass gain, changed bone morphology, and altered liver, muscle, and brain metabolism in ways indicative of a greater ability to utilize nutrients for anabolism. These effects were also documented in gnotobiotic piglets using the same consortium and Malawian diet. These preclinical models indicate a causal, microbiota-dependent relationship between S-BMO and growth promotion.


Assuntos
Desenvolvimento Infantil , Desnutrição/dietoterapia , Leite Humano/química , Leite/química , Oligossacarídeos/metabolismo , Animais , Bacteroides fragilis/genética , Bifidobacterium/classificação , Bifidobacterium/genética , Química Encefálica , Modelos Animais de Doenças , Escherichia coli/genética , Fezes/microbiologia , Vida Livre de Germes , Humanos , Lactente , Malaui , Masculino , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Microbiota
11.
Science ; 351(6275)2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26912898

RESUMO

Undernourished children exhibit impaired development of their gut microbiota. Transplanting microbiota from 6- and 18-month-old healthy or undernourished Malawian donors into young germ-free mice that were fed a Malawian diet revealed that immature microbiota from undernourished infants and children transmit impaired growth phenotypes. The representation of several age-discriminatory taxa in recipient animals correlated with lean body mass gain; liver, muscle, and brain metabolism; and bone morphology. Mice were cohoused shortly after receiving microbiota from healthy or severely stunted and underweight infants; age- and growth-discriminatory taxa from the microbiota of the former were able to invade that of the latter, which prevented growth impairments in recipient animals. Adding two invasive species, Ruminococcus gnavus and Clostridium symbiosum, to the microbiota from undernourished donors also ameliorated growth and metabolic abnormalities in recipient animals. These results provide evidence that microbiota immaturity is causally related to undernutrition and reveal potential therapeutic targets and agents.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal/fisiologia , Transtornos da Nutrição do Lactente/microbiologia , Animais , Bifidobacterium/fisiologia , Peso Corporal , Desenvolvimento Ósseo , Clostridiales/fisiologia , Modelos Animais de Doenças , Fezes/microbiologia , Fêmur/crescimento & desenvolvimento , Vida Livre de Germes , Humanos , Lactente , Transtornos da Nutrição do Lactente/metabolismo , Malaui , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...