Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 17: 568-580, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32258219

RESUMO

Phenylketonuria is an inborn error of metabolism caused by loss of function of the liver-expressed enzyme phenylalanine hydroxylase and is characterized by elevated systemic phenylalanine levels that are neurotoxic. Current therapies do not address the underlying genetic disease or restore the natural metabolic pathway resulting in the conversion of phenylalanine to tyrosine. A family of hepatotropic clade F adeno-associated viruses (AAVs) was isolated from human CD34+ hematopoietic stem cells (HSCs) and one (AAVHSC15) was utilized to deliver a vector to correct the phenylketonuria phenotype in Pahenu2 mice. The AAVHSC15 vector containing a codon-optimized form of the human phenylalanine hydroxylase cDNA was administered as a single intravenous dose to Pahenu2 mice maintained on a phenylalanine-containing normal chow diet. Optimization of the transgene resulted in a vector that produced a sustained reduction in serum phenylalanine and normalized tyrosine levels for the lifespan of Pahenu2 mice. Brain levels of phenylalanine and the downstream serotonin metabolite 5-hydroxyindoleacetic acid were restored. In addition, the coat color of treated mice darkened following treatment, indicating restoration of the phenylalanine metabolic pathway. Taken together, these data support the potential of an AAVHSC15-based gene therapy as an investigational therapeutic for phenylketonuria patients.

2.
Hum Vaccin Immunother ; 13(11): 2738-2741, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-28933665

RESUMO

There is a global shortage of equine-derived diphtheria anti-toxin (DAT) for diphtheria treatment. There are few existing data on serum antibody concentrations and neutralizing activity post-treatment to support development of new therapeutics. Antibody concentrations were quantified by ELISA and anti-toxin neutralizing activity by cytotoxicity assay in serum from 4 patients receiving DAT for suspected diphtheria. Using linear mixed effects modeling, estimated mean (SE) half-life was 78.2 (20.0) hours. Maximum serum neutralizing activity ranged from 28.42-38.64 AU/mL with an estimated mean AUC1-72 of 1396.7 (399.3) AU/mL*hr. These data provide a standard of comparison for development of novel anti-toxins to replace DAT.


Assuntos
Anticorpos Anti-Idiotípicos/sangue , Anticorpos Antibacterianos/imunologia , Antitoxina Diftérica/imunologia , Difteria/terapia , Administração Intravenosa , Adulto , Idoso , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Antibacterianos/administração & dosagem , Anticorpos Antibacterianos/metabolismo , Anticorpos Antibacterianos/uso terapêutico , Chlorocebus aethiops , Testes Imunológicos de Citotoxicidade , Difteria/sangue , Difteria/imunologia , Antitoxina Diftérica/administração & dosagem , Antitoxina Diftérica/metabolismo , Antitoxina Diftérica/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Feminino , Meia-Vida , Cavalos , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Células Vero
3.
Sci Rep ; 7: 41339, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128301

RESUMO

Progress in regenerative medicine requires reverse-engineering cellular control networks to infer perturbations with desired systems-level outcomes. Such dynamic models allow phenotypic predictions for novel perturbations to be rapidly assessed in silico. Here, we analyzed a Xenopus model of conversion of melanocytes to a metastatic-like phenotype only previously observed in an all-or-none manner. Prior in vivo genetic and pharmacological experiments showed that individual animals either fully convert or remain normal, at some characteristic frequency after a given perturbation. We developed a Machine Learning method which inferred a model explaining this complex, stochastic all-or-none dataset. We then used this model to ask how a new phenotype could be generated: animals in which only some of the melanocytes converted. Systematically performing in silico perturbations, the model predicted that a combination of altanserin (5HTR2 inhibitor), reserpine (VMAT inhibitor), and VP16-XlCreb1 (constitutively active CREB) would break the all-or-none concordance. Remarkably, applying the predicted combination of three reagents in vivo revealed precisely the expected novel outcome, resulting in partial conversion of melanocytes within individuals. This work demonstrates the capability of automated analysis of dynamic models of signaling networks to discover novel phenotypes and predictively identify specific manipulations that can reach them.


Assuntos
Melanócitos/metabolismo , Modelos Biológicos , Xenopus laevis/metabolismo , Animais , Simulação por Computador , Fenótipo , Pigmentação , Reprodutibilidade dos Testes , Transdução de Sinais
4.
Oncotarget ; 7(15): 19575-88, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26988909

RESUMO

It has long been known that the resting potential of tumor cells is depolarized relative to their normal counterparts. More recent work has provided evidence that resting potential is not just a readout of cell state: it regulates cell behavior as well. Thus, the ability to control resting potential in vivo would provide a powerful new tool for the study and treatment of tumors, a tool capable of revealing living-state physiological information impossible to obtain using molecular tools applied to isolated cell components. Here we describe the first use of optogenetics to manipulate ion-flux mediated regulation of membrane potential specifically to prevent and cause regression of oncogene-induced tumors. Injection of mutant-KRAS mRNA induces tumor-like structures with many documented similarities to tumors, in Xenopus tadpoles. We show that expression and activation of either ChR2D156A, a blue-light activated cation channel, or Arch, a green-light activated proton pump, both of which hyperpolarize cells, significantly lowers the incidence of KRAS tumor formation. Excitingly, we also demonstrate that activation of co-expressed light-activated ion translocators after tumor formation significantly increases the frequency with which the tumors regress in a process called normalization. These data demonstrate an optogenetic approach to dissect the biophysics of cancer. Moreover, they provide proof-of-principle for a novel class of interventions, directed at regulating cell state by targeting physiological regulators that can over-ride the presence of mutations.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/efeitos da radiação , Luz , Optogenética/métodos , Animais , Antineoplásicos/farmacologia , Proteínas Arqueais/genética , Transformação Celular Neoplásica/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Embrião não Mamífero/efeitos da radiação , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Potenciais da Membrana/efeitos da radiação , Mutação , Optogenética/instrumentação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rodopsina/genética , Xenopus laevis
5.
Sci Signal ; 8(397): ra99, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26443706

RESUMO

Experimentally induced depolarization of resting membrane potential in "instructor cells" in Xenopus laevis embryos causes hyperpigmentation in an all-or-none fashion in some tadpoles due to excess proliferation and migration of melanocytes. We showed that this stochastic process involved serotonin signaling, adenosine 3',5'-monophosphate (cAMP), and the transcription factors cAMP response element-binding protein (CREB), Sox10, and Slug. Transcriptional microarray analysis of embryos taken at stage 15 (early neurula) and stage 45 (free-swimming tadpole) revealed changes in the abundance of 45 and 517 transcripts, respectively, between control embryos and embryos exposed to the instructor cell-depolarizing agent ivermectin. Bioinformatic analysis revealed that the human homologs of some of the differentially regulated genes were associated with cancer, consistent with the induced arborization and invasive behavior of converted melanocytes. We identified a physiological circuit that uses serotonergic signaling between instructor cells, melanotrope cells of the pituitary, and melanocytes to control the proliferation, cell shape, and migration properties of the pigment cell pool. To understand the stochasticity and properties of this multiscale signaling system, we applied a computational machine-learning method that iteratively explored network models to reverse-engineer a stochastic dynamic model that recapitulated the frequency of the all-or-none hyperpigmentation phenotype produced in response to various pharmacological and molecular genetic manipulations. This computational approach may provide insight into stochastic cellular decision-making that occurs during normal development and pathological conditions, such as cancer.


Assuntos
Simulação por Computador , Hiperpigmentação/fisiopatologia , Larva/fisiologia , Melanócitos/fisiologia , Modelos Biológicos , Serotonina/fisiologia , Pigmentação da Pele/fisiologia , Xenopus laevis/fisiologia , Animais , Proteína de Ligação a CREB/fisiologia , Movimento Celular , Forma Celular , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Ivermectina/farmacologia , Aprendizado de Máquina , Melanócitos/citologia , Potenciais da Membrana/efeitos dos fármacos , Hipófise/fisiologia , Fatores de Transcrição SOXE/fisiologia , Transdução de Sinais/fisiologia , Processos Estocásticos , Fatores de Transcrição/fisiologia , Transcrição Gênica , Proteínas de Xenopus/fisiologia , Xenopus laevis/embriologia
6.
Int J Dev Biol ; 59(7-9): 303-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26198143

RESUMO

The correct anatomical placement and precise determination of specific cell types is required for the establishment of normal embryonic patterning. Understanding these processes is also important for progress in regenerative medicine and cancer biology. Transmembrane voltage gradients across embryonic tissues can mediate cellular communication to regulate the processes of proliferation, migration, and differentiation. Our past work showed that selective depolarization of an endogenous instructor cell population in Xenopus laevis in vivo induced a melanoma-like phenotype in the absence of genetic damage. Here, we use a hypersensitive glycine-gated chloride channel (GlyR) under control of tissue-specific promoters to show that instructor cells resident within muscle are more effective at triggering the metastatic conversion of ectodermal melanocytes than those similar cells within the nervous system. Moreover, depolarization of muscle cells results in aberrant muscle patterning and the appearance of cells expressing muscle markers within the neural tube, which impacts but does not abolish the animals' ability to learn in an associative conditioning assay. Taken together, our data reveal new details of long-range (non-cell-autonomous) reprogramming of cell behavior via alteration of the resting potential of specific embryonic subpopulations.


Assuntos
Melanócitos/fisiologia , Potenciais da Membrana/fisiologia , Células Musculares/fisiologia , Desenvolvimento Muscular/fisiologia , Animais , Diferenciação Celular/fisiologia , Melanócitos/citologia , Xenopus laevis
7.
Phys Biol ; 9(6): 065002, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23196890

RESUMO

Cancer may result from localized failure of instructive cues that normally orchestrate cell behaviors toward the patterning needs of the organism. Steady-state gradients of transmembrane voltage (V(mem)) in non-neural cells are instructive, epigenetic signals that regulate pattern formation during embryogenesis and morphostatic repair. Here, we review molecular data on the role of bioelectric cues in cancer and present new findings in the Xenopus laevis model on how the microenvironment's biophysical properties contribute to cancer in vivo. First, we investigated the melanoma-like phenotype arising from serotonergic signaling by 'instructor' cells-a cell population that is able to induce a metastatic phenotype in normal melanocytes. We show that when these instructor cells are depolarized, blood vessel patterning is disrupted in addition to the metastatic phenotype induced in melanocytes. Surprisingly, very few instructor cells need to be depolarized for the hyperpigmentation phenotype to occur; we present a model of antagonistic signaling by serotonin receptors that explains the unusual all-or-none nature of this effect. In addition to the body-wide depolarization-induced metastatic phenotype, we investigated the bioelectrical properties of tumor-like structures induced by canonical oncogenes and cancer-causing compounds. Exposure to carcinogen 4-nitroquinoline 1-oxide (4NQO) induces localized tumors, but has a broad (and variable) effect on the bioelectric properties of the whole body. Tumors induced by oncogenes show aberrantly high sodium content, representing a non-invasive diagnostic modality. Importantly, depolarized transmembrane potential is not only a marker of cancer but is functionally instructive: susceptibility to oncogene-induced tumorigenesis is significantly reduced by forced prior expression of hyperpolarizing ion channels. Importantly, the same effect can be achieved by pharmacological manipulation of endogenous chloride channels, suggesting a strategy for cancer suppression that does not require gene therapy. Together, these data extend our understanding of the recently demonstrated role of transmembrane potential in tumor formation and metastatic cell behavior. V(mem) is an important non-genetic biophysical aspect of the microenvironment that regulates the balance between normally patterned growth and carcinogenesis.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Potenciais da Membrana , Oncogenes , Microambiente Tumoral , 4-Nitroquinolina-1-Óxido/toxicidade , Animais , Vasos Sanguíneos/embriologia , Padronização Corporal , Carcinógenos/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/patologia , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/induzido quimicamente , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Transdução de Sinais , Sódio/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/fisiologia
8.
Proc Natl Acad Sci U S A ; 109(31): 12586-91, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-22802643

RESUMO

Many types of embryos' bodyplans exhibit consistently oriented laterality of the heart, viscera, and brain. Errors of left-right patterning present an important class of human birth defects, and considerable controversy exists about the nature and evolutionary conservation of the molecular mechanisms that allow embryos to reliably orient the left-right axis. Here we show that the same mutations in the cytoskeletal protein tubulin that alter asymmetry in plants also affect very early steps of left-right patterning in nematode and frog embryos, as well as chirality of human cells in culture. In the frog embryo, tubulin α and tubulin γ-associated proteins are required for the differential distribution of maternal proteins to the left or right blastomere at the first cell division. Our data reveal a remarkable molecular conservation of mechanisms initiating left-right asymmetry. The origin of laterality is cytoplasmic, ancient, and highly conserved across kingdoms, a fundamental feature of the cytoskeleton that underlies chirality in cells and multicellular organisms.


Assuntos
Blastômeros/metabolismo , Padronização Corporal/fisiologia , Divisão Celular/fisiologia , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Blastômeros/citologia , Células HL-60 , Humanos , Xenopus laevis
9.
Dis Model Mech ; 4(1): 67-85, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20959630

RESUMO

Understanding the mechanisms that coordinate stem cell behavior within the host is a high priority for developmental biology, regenerative medicine and oncology. Endogenous ion currents and voltage gradients function alongside biochemical cues during pattern formation and tumor suppression, but it is not known whether bioelectrical signals are involved in the control of stem cell progeny in vivo. We studied Xenopus laevis neural crest, an embryonic stem cell population that gives rise to many cell types, including melanocytes, and contributes to the morphogenesis of the face, heart and other complex structures. To investigate how depolarization of transmembrane potential of cells in the neural crest's environment influences its function in vivo, we manipulated the activity of the native glycine receptor chloride channel (GlyCl). Molecular-genetic depolarization of a sparse, widely distributed set of GlyCl-expressing cells non-cell-autonomously induces a neoplastic-like phenotype in melanocytes: they overproliferate, acquire an arborized cell shape and migrate inappropriately, colonizing numerous tissues in a metalloprotease-dependent fashion. A similar effect was observed in human melanocytes in culture. Depolarization of GlyCl-expressing cells induces these drastic changes in melanocyte behavior via a serotonin-transporter-dependent increase of extracellular serotonin (5-HT). These data reveal GlyCl as a molecular marker of a sparse and heretofore unknown cell population with the ability to specifically instruct neural crest derivatives, suggest transmembrane potential as a tractable signaling modality by which somatic cells can control stem cell behavior at considerable distance, identify a new biophysical aspect of the environment that confers a neoplastic-like phenotype upon stem cell progeny, reveal a pre-neural role for serotonin and its transporter, and suggest a novel strategy for manipulating stem cell behavior.


Assuntos
Transformação Celular Neoplásica/patologia , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/patologia , Potenciais da Membrana , Receptores de Glicina/metabolismo , Serotonina/metabolismo , Animais , Contagem de Células , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Forma Celular/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Cloretos/metabolismo , Coristoma/patologia , Epiderme/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hiperpigmentação/metabolismo , Hiperpigmentação/patologia , Ativação do Canal Iônico/efeitos dos fármacos , Ivermectina/farmacologia , Melanócitos/efeitos dos fármacos , Melanoma/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Modelos Biológicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Glicina/genética , Transdução de Sinais/efeitos dos fármacos , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...