Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Med Virol ; 96(7): e29782, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39011762

RESUMO

Extracellular vesicles (EVs) are shown to be a novel viral transmission model capable of increasing a virus's tropism. According to our earlier research, cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or transfected with envelope protein plasmids generate a novel type of EVs that are micrometer-sized and able to encase virus particles. Here, we showed the capacity of these EVs to invade various animals both in vitro and in vivo independent of the angiotensin-converting enzyme 2 receptor. First, via macropinocytosis, intact EVs produced from Vero E6 (monkey) cells were able to enter cells from a variety of animals, including cats, dogs, bats, hamsters, and minks, and vice versa. Second, when given to zebrafish with cutaneous wounds, the EVs showed favorable stability in aqueous environments and entered the fish. Moreover, infection of wild-type (WT) mice with heterogeneous EVs carrying SARS-CoV-2 particles led to a strong cytokine response and a notable amount of lung damage. Conversely, free viral particles did not infect WT mice. These results highlight the variety of processes behind viral transmission and cross-species evolution by indicating that EVs may be possible vehicles for SARS-CoV-2 spillover and raising risk concerns over EVs' potential for viral gene transfer.


Assuntos
COVID-19 , Vesículas Extracelulares , SARS-CoV-2 , Animais , Vesículas Extracelulares/virologia , Vesículas Extracelulares/metabolismo , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , SARS-CoV-2/genética , COVID-19/transmissão , COVID-19/virologia , Camundongos , Chlorocebus aethiops , Células Vero , Humanos , Cricetinae , Proteínas do Envelope de Coronavírus/metabolismo , Proteínas do Envelope de Coronavírus/genética , Cães , Peixe-Zebra/virologia , Gatos , Quirópteros/virologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética
2.
PLoS Biol ; 10(8): e1001374, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22904685

RESUMO

The brain blood vasculature consists of a highly ramified vessel network that is tailored to meet its physiological functions. How the brain vasculature is formed has long been fascinating biologists. Here we report that the developing vasculature in the zebrafish midbrain undergoes not only angiogenesis but also extensive vessel pruning, which is driven by changes in blood flow. This pruning process shapes the initial exuberant interconnected meshwork into a simplified architecture. Using in vivo long-term serial confocal imaging of the same zebrafish larvae during 1.5-7.5 d post-fertilization, we found that the early formed midbrain vasculature consisted of many vessel loops and higher order segments. Vessel pruning occurred preferentially at loop-forming segments via a process mainly involving lateral migration of endothelial cells (ECs) from pruned to unpruned segments rather than EC apoptosis, leading to gradual reduction in the vasculature complexity with development. Compared to unpruned ones, pruned segments exhibited a low and variable blood flow, which further decreased irreversibly prior to the onset of pruning. Local blockade of blood flow with micro-bead obstruction led to vessel pruning, whereas increasing blood flow by noradrenergic elevation of heartbeat impeded the pruning process. Furthermore, the occurrence of vessel pruning could be largely predicted by haemodynamics-based numerical simulation of vasculature refinement. Thus, changes of blood flow drive vessel pruning via lateral migration of ECs, leading to the simplification of the vasculature and possibly efficient routing of blood flow in the developing brain.


Assuntos
Hemodinâmica , Mesencéfalo/irrigação sanguínea , Neovascularização Fisiológica , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados/fisiologia , Velocidade do Fluxo Sanguíneo , Movimento Celular , Embrião não Mamífero/irrigação sanguínea , Embrião não Mamífero/embriologia , Embrião não Mamífero/fisiologia , Desenvolvimento Embrionário , Células Endoteliais/fisiologia , Larva/fisiologia , Macrófagos/fisiologia , Mesencéfalo/anatomia & histologia , Mesencéfalo/fisiologia , Microscopia Confocal/métodos , Modelos Biológicos , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/genética
3.
Angew Chem Int Ed Engl ; 54(47): 14026-30, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26228648

RESUMO

Multifunctional stimuli-responsive nanotheranostic systems are highly desirable for realizing simultaneous biomedical imaging and on-demand therapy with minimized adverse effects. Herein, we present the construction of an intelligent X-ray-controlled NO-releasing upconversion nanotheranostic system (termed as PEG-USMSs-SNO) by engineering UCNPs with S-nitrosothiol (R-SNO)-grafted mesoporous silica. The PEG-USMSs-SNO is designed to respond sensitively to X-ray radiation for breaking down the S-N bond of SNO to release NO, which leads to X-ray dose-controlled NO release for on-demand hypoxic radiosensitization besides upconversion luminescent imaging through UCNPs in vitro and in vivo. Thanks to the high live-body permeability of X-ray, our developed PEG-USMSs-SNO may provide a new technique for achieving depth-independent controlled NO release and positioned radiotherapy enhancement against deep-seated solid tumors.


Assuntos
Nanopartículas/química , Nanopartículas/efeitos da radiação , Óxido Nítrico/química , Nanomedicina Teranóstica/métodos , Raios X
4.
J Am Chem Soc ; 136(27): 9701-9, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-24956326

RESUMO

Hypoxia is a distinct feature of malignant solid tumors, which is a possible causative factor for the serious resistance to chemo- and radiotherapy or the development of invasion and metastasis. The exploration of nanosensors with the capabilities like the accurate diagnosis of hypoxic level will be helpful to estimate the malignant degree of tumor and subsequently implement more effective personalized treatment. Here, we report the design and synthesis of nanosensors that can selectively and reversibly detect the level of hypoxia both in vitro and in vivo. The designed nanosensor is composed of two main moieties: oxygen indicator [Ru(dpp)3](2+)Cl2 for detection of hypoxia and upconversion nanoparticles for offering the excitation light of [Ru(dpp)3](2+)Cl2 by upconversion process under 980 nm exposure. The results show that the nanosensors can reversibly become quenched or luminescent under hyperoxic or hypoxic conditions, respectively. Compared with free [Ru(dpp)3](2+)Cl2, the designed nanosensors exhibit enhanced sensitivity for the detection of oxygen in hypoxic regions. More attractively, the nanosensors can image hypoxic regions with high penetration depth because the absorption and emission wavelength are within the NIR and far-red region, respectively. Most importantly, nanosensors display a high selectivity for detection of relevant oxygen changes in cells and zebrafish.


Assuntos
Hipóxia , Nanopartículas/química , Nanotecnologia/instrumentação , Compostos Organometálicos/química , Oxigênio/análise , Rutênio/química , Adsorção , Animais , Linhagem Celular Tumoral , Humanos , Microscopia Confocal , Estrutura Molecular , Compostos Organometálicos/síntese química , Tamanho da Partícula , Propriedades de Superfície , Peixe-Zebra
5.
Angew Chem Int Ed Engl ; 53(18): 4551-5, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24668766

RESUMO

Insufficient or excess drug doses, due to unknown actual drug concentrations at the focus, are one of the main causes of chemotherapy failure for cancers. In this regard, the real-time monitoring of the release of anticancer drugs from nanoparticle drug delivery systems is of crucial importance, but it remains a critical and unsolved challenge. Herein, we report the proposal and development of a novel concept of real-time monitoring of NIR-triggered drug release in vitro and in vivo by using simultaneous upconverted luminescence (UCL) and magnetic resonance (MR) imaging. Such a monitoring strategy features the high sensitivity of UCL and the high-resolution, noninvasiveness, and tissue-depth-independence of MR imaging. The dual-mode real-time and quantitative monitoring of drug release can be applied to determine online the drug concentrations in vivo in the tissue regions of interest and, therefore, to avoid insufficient or excess drug dosings.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacocinética , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Medições Luminescentes/métodos , Espectroscopia de Ressonância Magnética/métodos , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Gadolínio/química , Células HeLa , Humanos , Nanocompostos/química , Nanopartículas/química , Distribuição Tecidual
6.
Cell Rep ; 43(1): 113652, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-38175750

RESUMO

Blood flow is known to regulate cerebrovascular development through acting on vascular endothelial cells (ECs). As an indispensable component of the neurovascular unit, brain pericytes physically couple with ECs and play vital roles in blood-brain barrier integrity maintenance and neurovascular coupling. However, it remains unclear whether blood flow affects brain pericyte development. Using in vivo time-lapse imaging of larval zebrafish, we monitored the developmental dynamics of brain pericytes and found that they proliferate to expand their population and increase their coverage to brain vessels. In combination with pharmacological and genetic approaches, we demonstrated that blood flow enhances brain pericyte proliferation through Piezo1 expressed in ECs. Moreover, we identified that EC-intrinsic Notch signaling is downstream of Piezo1 to promote the activation of Notch signaling in pericytes. Thus, our findings reveal a role of blood flow in pericyte proliferation, extending the functional spectrum of hemodynamics on cerebrovascular development.


Assuntos
Pericitos , Peixe-Zebra , Animais , Células Endoteliais/fisiologia , Encéfalo/fisiologia , Barreira Hematoencefálica , Hemodinâmica , Proliferação de Células , Canais Iônicos , Proteínas de Peixe-Zebra
7.
Circ Res ; 106(7): 1221-32, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20185799

RESUMO

RATIONALE: Wiring vascular and neural networks are known to share common molecular signaling pathways. Activation of transient receptor potential type C channels (TRPCs) has recently been shown to underlie chemotropic guidance of neural axons. It is thus of interest to examine whether TRPCs are also involved in vascular development. OBJECTIVE: To determine the role of TRPC1 in angiogenesis in vivo during zebrafish development. METHODS AND RESULTS: Knockdown of zebrafish trpc1 by antisense morpholino oligonucleotides severely disrupted angiogenic sprouting of intersegmental vessels (ISVs) in zebrafish larvae. This angiogenic defect was prevented by overexpression of a morpholino oligonucleotide-resistant form of zebrafish trpc1 mRNA. Cell transplantation analysis showed that this requirement of Trpc1 for ISV growth was endothelial cell-autonomous. In vivo time-lapse imaging further revealed that the angiogenic defect was attributable to impairment of filopodia extension, migration, and proliferation of ISV tip cells. Furthermore, Trpc1 acted synergistically with vascular endothelial growth factor A (Vegf-a) in controlling ISV growth, and appeared to be downstream to Vegf-a in controlling angiogenesis, as evidence by the findings that Trpc1 was required for Vegf-a-induced ectopic angiogenesis of subintestinal veins and phosphorylation of extracellular signal-regulated kinase. CONCLUSIONS: These results provide the first in vivo evidence that TRPC1 is essential for angiogenesis, reminiscent of the role of TRPCs in axon guidance. It implicates that TRPC1 may represent a potential target for treating pathological angiogenesis.


Assuntos
Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Canais de Cátion TRPC/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Vasos Sanguíneos/embriologia , Movimento Celular , Proliferação de Células , Células Endoteliais/transplante , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Microscopia Confocal , Microscopia de Vídeo , Oligonucleotídeos Antissenso/metabolismo , Fosforilação , Pseudópodes/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Canais de Cátion TRPC/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética
8.
Anesthesiology ; 114(2): 391-400, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21245739

RESUMO

BACKGROUND: Celecoxib, a cyclooxygenase-2 inhibitor, is a commonly ingested drug that is used by some women during pregnancy. Although use of celecoxib is associated with increased cardiovascular risk in adults, its effect on fetal heart development remains unknown. METHODS: Zebrafish embryos were exposed to celecoxib or other relevant drugs from tailbud stage (10.3-72 h postfertilization). Heart looping and valve formation were examined at different developmental stages by in vivo confocal imaging. In addition, whole mount in situ hybridization was performed to examine drug-induced changes in the expression of heart valve marker genes. RESULTS: In celecoxib-treated zebrafish embryos, the heart failed to undergo normal looping and the heart valve was absent, causing serious blood regurgitation. Furthermore, celecoxib treatment disturbed the restricted expression of the heart valve markers bone morphogenetic protein 4 and versican-but not the cardiac chamber markers cardiac myosin light chain 2, ventricular myosin heavy chain, and atrial myosin heavy chain. These defects in heart development were markedly relieved by treatment with the cyclooxygenase-2 downstream product prostaglandin E2, and mimicked by the cyclooxygenase-2 inhibitor NS398, implying that celecoxib-induced heart defects were caused by the inhibition of cyclooxygenase-2 activity. CONCLUSIONS: These findings provide the first in vivo evidence that celecoxib exposure impairs heart development in zebrafish embryos by inhibiting cyclooxygenase-2 activity.


Assuntos
Inibidores de Ciclo-Oxigenase 2/efeitos adversos , Ciclo-Oxigenase 2/efeitos dos fármacos , Cardiopatias/induzido quimicamente , Cardiopatias/embriologia , Pirazóis/efeitos adversos , Sulfonamidas/efeitos adversos , Acetaminofen/administração & dosagem , Analgésicos não Narcóticos/administração & dosagem , Análise de Variância , Animais , Celecoxib , Inibidores de Ciclo-Oxigenase 2/administração & dosagem , Relação Dose-Resposta a Droga , Coração/efeitos dos fármacos , Coração/embriologia , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/embriologia , Pirazóis/administração & dosagem , Sulfonamidas/administração & dosagem , Peixe-Zebra
9.
Sci Adv ; 6(16): eaax9757, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32494594

RESUMO

Potassium ion (K+) concentration fluctuates in various biological processes. A number of K+ probes have been developed to monitor such fluctuations through optical imaging. However, the currently available K+ probes are far from being sensitive enough in detecting physiological fluctuations in living animals. Furthermore, the monitoring of deep tissues is not applicable because of short-wavelength excitation prevailingly used so far. Here, we report a highly sensitive and selective nanosensor for near-infrared (NIR) K+ imaging in living cells and animals. The nanosensor is constructed by encapsulating upconversion nanoparticles (UCNPs) and a commercial K+ indicator in the hollow cavity of mesoporous silica nanoparticles, followed by coating a K+-selective filter membrane. The membrane adsorbs K+ from the medium and filters out interfering cations. The UCNPs convert NIR to ultraviolet light, which excites the K+ indicator, thus allowing the detection of the fluctuations of K+ concentration in cultured cells and intact mouse brains.

10.
Cell Res ; 27(7): 882-897, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28429770

RESUMO

Vascular integrity helps maintain brain microenvironment homeostasis, which is critical for the normal development and function of the central nervous system. It is known that neural cells can regulate brain vascular integrity. However, due to the high complexity of neurovascular interactions involved, understanding of the neural regulation of brain vascular integrity is still rudimentary. Using intact zebrafish larvae and cultured rodent brain cells, we find that neurons transfer miR-132, a highly conserved and neuron-enriched microRNA, via secreting exosomes to endothelial cells (ECs) to maintain brain vascular integrity. Following translocation to ECs through exosome internalization, miR-132 regulates the expression of vascular endothelial cadherin (VE-cadherin), an important adherens junction protein, by directly targeting eukaryotic elongation factor 2 kinase (eef2k). Disruption of neuronal miR-132 expression or exosome secretion, or overexpression of vascular eef2k impairs VE-cadherin expression and brain vascular integrity. Our study indicates that miR-132 acts as an intercellular signal mediating neural regulation of the brain vascular integrity and suggests that the neuronal exosome is a novel avenue for neurovascular communication.


Assuntos
Encéfalo/irrigação sanguínea , Exossomos/metabolismo , Hemorragias Intracranianas/metabolismo , MicroRNAs/metabolismo , Neurônios/metabolismo , Análise de Variância , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Exossomos/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Hemorragias Intracranianas/patologia , Larva , Camundongos , MicroRNAs/genética , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Peixe-Zebra
12.
Oncotarget ; 6(20): 17891-4, 2015 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-26143640

RESUMO

The TALEN and CRISPR/Cas9 nuclease systems have been extensively utilized in genomic engineering of model organisms. In zebrafish, the nuclease systems have been successfully applied in generating loss-of-function knockout lines. However, genome-specific knockin techniques in zebrafish are still at the beginning. In this perspective, we briefly summarize the recent progresses on knockin approaches in zebrafish with a special focus on the newly developed intron-based knockin method.


Assuntos
Técnicas de Introdução de Genes , Íntrons , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Reparo do DNA por Junção de Extremidades , Regulação da Expressão Gênica , Genes Reporter , Genótipo , Fenótipo , Reparo de DNA por Recombinação , Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA