Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(4): 286, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38653992

RESUMO

The progression of human degenerative and hypoxic/ischemic diseases is accompanied by widespread cell death. One death process linking iron-catalyzed reactive species with lipid peroxidation is ferroptosis, which shows hallmarks of both programmed and necrotic death in vitro. While evidence of ferroptosis in neurodegenerative disease is indicated by iron accumulation and involvement of lipids, a stable marker for ferroptosis has not been identified. Its prevalence is thus undetermined in human pathophysiology, impeding recognition of disease areas and clinical investigations with candidate drugs. Here, we identified ferroptosis marker antigens by analyzing surface protein dynamics and discovered a single protein, Fatty Acid-Binding Protein 5 (FABP5), which was stabilized at the cell surface and specifically elevated in ferroptotic cell death. Ectopic expression and lipidomics assays demonstrated that FABP5 drives redistribution of redox-sensitive lipids and ferroptosis sensitivity in a positive-feedback loop, indicating a role as a functional biomarker. Notably, immunodetection of FABP5 in mouse stroke penumbra and in hypoxic postmortem patients was distinctly associated with hypoxically damaged neurons. Retrospective cell death characterized here by the novel ferroptosis biomarker FABP5 thus provides first evidence for a long-hypothesized intrinsic ferroptosis in hypoxia and inaugurates a means for pathological detection of ferroptosis in tissue.


Assuntos
Biomarcadores , Proteínas de Ligação a Ácido Graxo , Ferroptose , Proteínas de Neoplasias , Proteínas de Ligação a Ácido Graxo/metabolismo , Animais , Humanos , Biomarcadores/metabolismo , Camundongos , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Camundongos Endogâmicos C57BL , Peroxidação de Lipídeos , Masculino
2.
Cancers (Basel) ; 15(21)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37958423

RESUMO

Glioblastoma is the most common primary brain cancer in adults and represents one of the worst cancer diagnoses for patients. Suffering from a poor prognosis and limited treatment options, tumor recurrences are virtually inevitable. Additionally, treatment resistance is very common for this disease and worsens the prognosis. These and other factors are hypothesized to be largely due to the fact that glioblastoma cells are known to be able to obtain stem-like traits, thereby driving these phenotypes. Recently, we have shown that the in vitro and ex vivo treatment of glioblastoma stem-like cells with the hormonally active form of vitamin D3, calcitriol (1α,25(OH)2-vitamin D3) can block stemness in a subset of cell lines and reduce tumor growth. Here, we expanded our cell panel to over 40 different cultures and can show that, while half of the tested cell lines are sensitive, a quarter can be classified as high responders. Using genetic and proteomic analysis, we further determined that treatment success can be partially explained by specific polymorphism of the vitamin D3 receptor and that high responders display a proteome suggestive of blockade of stemness, as well as migratory potential.

3.
J Extracell Vesicles ; 12(2): e12305, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36775986

RESUMO

Extracellular vesicles (EVs) carry diverse bioactive components including nucleic acids, proteins, lipids and metabolites that play versatile roles in intercellular and interorgan communication. The capability to modulate their stability, tissue-specific targeting and cargo render EVs as promising nanotherapeutics for treating heart, lung, blood and sleep (HLBS) diseases. However, current limitations in large-scale manufacturing of therapeutic-grade EVs, and knowledge gaps in EV biogenesis and heterogeneity pose significant challenges in their clinical application as diagnostics or therapeutics for HLBS diseases. To address these challenges, a strategic workshop with multidisciplinary experts in EV biology and U.S. Food and Drug Administration (USFDA) officials was convened by the National Heart, Lung and Blood Institute. The presentations and discussions were focused on summarizing the current state of science and technology for engineering therapeutic EVs for HLBS diseases, identifying critical knowledge gaps and regulatory challenges and suggesting potential solutions to promulgate translation of therapeutic EVs to the clinic. Benchmarks to meet the critical quality attributes set by the USFDA for other cell-based therapeutics were discussed. Development of novel strategies and approaches for scaling-up EV production and the quality control/quality analysis (QC/QA) of EV-based therapeutics were recognized as the necessary milestones for future investigations.


Assuntos
Vesículas Extracelulares , Ácidos Nucleicos , Estados Unidos , Vesículas Extracelulares/metabolismo , Comunicação Celular , Ácidos Nucleicos/metabolismo , Pulmão/metabolismo , Sono
4.
Life Sci Alliance ; 4(12)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34663679

RESUMO

Extracellular vesicles (EVs) mediate intercellular signaling by transferring their cargo to recipient cells, but the functional consequences of signaling are not fully appreciated. RBC-derived EVs are abundant in circulation and have been implicated in regulating immune responses. Here, we use a transgenic mouse model for fluorescence-based mapping of RBC-EV recipient cells to assess the role of this intercellular signaling mechanism in heart disease. Using fluorescent-based mapping, we detected an increase in RBC-EV-targeted cardiomyocytes in a murine model of ischemic heart failure. Single cell nuclear RNA sequencing of the heart revealed a complex landscape of cardiac cells targeted by RBC-EVs, with enrichment of genes implicated in cell proliferation and stress signaling pathways compared with non-targeted cells. Correspondingly, cardiomyocytes targeted by RBC-EVs more frequently express cellular markers of DNA synthesis, suggesting the functional significance of EV-mediated signaling. In conclusion, our mouse model for mapping of EV-recipient cells reveals a complex cellular network of RBC-EV-mediated intercellular communication in ischemic heart failure and suggests a functional role for this mode of intercellular signaling.


Assuntos
Eritrócitos/metabolismo , Vesículas Extracelulares/metabolismo , Insuficiência Cardíaca/sangue , Infarto do Miocárdio/sangue , Miocárdio/metabolismo , RNA Nuclear/genética , RNA-Seq/métodos , Transdução de Sinais/genética , Análise de Célula Única/métodos , Animais , Comunicação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Voluntários Saudáveis , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo
5.
J Extracell Vesicles ; 10(12): e12159, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34664784

RESUMO

The intestinal microbiota influences mammalian host physiology in health and disease locally in the gut but also in organs devoid of direct contact with bacteria such as the liver and brain. Extracellular vesicles (EVs) or outer membrane vesicles (OMVs) released by microbes are increasingly recognized for their potential role as biological shuttle systems for inter-kingdom communication. However, physiologically relevant evidence for the transfer of functional biomolecules from the intestinal microbiota to individual host cells by OMVs in vivo is scarce. By introducing Escherichia coli engineered to express Cre-recombinase (E. coliCre ) into mice with a Rosa26.tdTomato-reporter background, we leveraged the Cre-LoxP system to report the transfer of bacterial OMVs to recipient cells in vivo. Colonizing the intestine of these mice with E. coliCre , resulted in Cre-recombinase induced fluorescent reporter gene-expression in cells along the intestinal epithelium, including intestinal stem cells as well as mucosal immune cells such as macrophages. Furthermore, even far beyond the gut, bacterial-derived Cre induced extended marker gene expression in a wide range of host tissues, including the heart, liver, kidney, spleen, and brain. Together, our findings provide a method and proof of principle that OMVs can serve as a biological shuttle system for the horizontal transfer of functional biomolecules between bacteria and mammalian host cells.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Escherichia coli/metabolismo , Microbioma Gastrointestinal/genética , Animais , Camundongos
6.
Nat Methods ; 18(9): 1013-1026, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34446922

RESUMO

Extracellular vesicles (EVs) are nano-sized lipid bilayer vesicles released by virtually every cell type. EVs have diverse biological activities, ranging from roles in development and homeostasis to cancer progression, which has spurred the development of EVs as disease biomarkers and drug nanovehicles. Owing to the small size of EVs, however, most studies have relied on isolation and biochemical analysis of bulk EVs separated from biofluids. Although informative, these approaches do not capture the dynamics of EV release, biodistribution, and other contributions to pathophysiology. Recent advances in live and high-resolution microscopy techniques, combined with innovative EV labeling strategies and reporter systems, provide new tools to study EVs in vivo in their physiological environment and at the single-vesicle level. Here we critically review the latest advances and challenges in EV imaging, and identify urgent, outstanding questions in our quest to unravel EV biology and therapeutic applications.


Assuntos
Vesículas Extracelulares , Microscopia/métodos , Animais , Corantes/química , Epitopos , Vesículas Extracelulares/química , Vesículas Extracelulares/patologia , Vesículas Extracelulares/fisiologia , Corantes Fluorescentes/química , Humanos
7.
Curr Opin Genet Dev ; 70: 61-65, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34153928

RESUMO

Recovery of brain function lost to disease or in old age is a challenging task in regenerative medicine. In the last two decades, therapeutic strategies have undergone significant shifts by a succession of major discoveries from adult neural stem cells and neurogenesis to the development of induced pluripotent stem cells to technologies for reprogramming cells in vitro and in vivo. Now, extracellular vesicles, small membrane-bound vesicles released by all cells and containing lipids, proteins, and nucleic acids, emerge as the next major technological opportunity. While substantial progress has been made on their potential use in therapy and EVs have entered many clinical trials, major aspects of their physiological role, in particular regarding their influence on brain function, remain unknown. However, a better understanding of their actual in vivo function, scope of communication, and possibilities to alter cellular processes in target cells will be needed. This review places EVs in the developing landscape of strategies for cellular repair of the brain and highlights their potential by looking at some recent progress in our understanding of their function in vivo.


Assuntos
Encéfalo/fisiopatologia , Vesículas Extracelulares/metabolismo , Medicina Regenerativa , Animais , Humanos
8.
PLoS Biol ; 18(3): e3000643, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32176686

RESUMO

Communication with the hematopoietic system is a vital component of regulating brain function in health and disease. Traditionally, the major routes considered for this neuroimmune communication are by individual molecules such as cytokines carried by blood, by neural transmission, or, in more severe pathologies, by the entry of peripheral immune cells into the brain. In addition, functional mRNA from peripheral blood can be directly transferred to neurons via extracellular vesicles (EVs), but the parameters that determine their uptake are unknown. Using varied animal models that stimulate neuronal activity by peripheral inflammation, optogenetics, and selective proteasome inhibition of dopaminergic (DA) neurons, we show that the transfer of EVs from blood is triggered by neuronal activity in vivo. Importantly, this transfer occurs not only in pathological stimulation but also by neuronal activation caused by the physiological stimulus of novel object placement. This discovery suggests a continuous role of EVs under pathological conditions as well as during routine cognitive tasks in the healthy brain.


Assuntos
Células Sanguíneas/citologia , Encéfalo/metabolismo , Vesículas Extracelulares/metabolismo , Inflamação/metabolismo , Animais , Células Sanguíneas/metabolismo , Encéfalo/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Feminino , Hipocampo/fisiologia , Inflamação/induzido quimicamente , Ácido Caínico/farmacologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos Transgênicos , Optogenética , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Técnicas Estereotáxicas , Ubiquitina/metabolismo
9.
EMBO J ; 37(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29903919

RESUMO

Glioblastoma is the most common and aggressive brain tumor, with a subpopulation of stem-like cells thought to mediate its recurring behavior and therapeutic resistance. The epithelial-mesenchymal transition (EMT) inducing factor Zeb1 was linked to tumor initiation, invasion, and resistance to therapy in glioblastoma, but how Zeb1 functions at molecular level and what genes it regulates remain poorly understood. Contrary to the common view that EMT factors act as transcriptional repressors, here we show that genome-wide binding of Zeb1 associates with both activation and repression of gene expression in glioblastoma stem-like cells. Transcriptional repression requires direct DNA binding of Zeb1, while indirect recruitment to regulatory regions by the Wnt pathway effector Lef1 results in gene activation, independently of Wnt signaling. Amongst glioblastoma genes activated by Zeb1 are predicted mediators of tumor cell migration and invasion, including the guanine nucleotide exchange factor Prex1, whose elevated expression is predictive of shorter glioblastoma patient survival. Prex1 promotes invasiveness of glioblastoma cells in vivo highlighting the importance of Zeb1/Lef1 gene regulatory mechanisms in gliomagenesis.


Assuntos
Glioblastoma/genética , Glioblastoma/patologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Via de Sinalização Wnt/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal/genética , Glioblastoma/mortalidade , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Invasividade Neoplásica/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
10.
JCI Insight ; 3(7)2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29618663

RESUMO

Extracellular RNA (exRNA) has emerged as an important transducer of intercellular communication. Advancing exRNA research promises to revolutionize biology and transform clinical practice. Recent efforts have led to cutting-edge research and expanded knowledge of this new paradigm in cell-to-cell crosstalk; however, gaps in our understanding of EV heterogeneity and exRNA diversity pose significant challenges for continued development of exRNA diagnostics and therapeutics. To unravel this complexity, the NIH convened expert teams to discuss the current state of the science, define the significant bottlenecks, and brainstorm potential solutions across the entire exRNA research field. The NIH Strategic Workshop on Extracellular RNA Transport helped identify mechanistic and clinical research opportunities for exRNA biology and provided recommendations on high priority areas of research that will advance the exRNA field.


Assuntos
Comunicação Celular/genética , Espaço Extracelular/metabolismo , Regulação da Expressão Gênica/imunologia , RNA/metabolismo , Animais , Comunicação Celular/imunologia , Congressos como Assunto , Modelos Animais de Doenças , Espaço Extracelular/genética , Espaço Extracelular/imunologia , Humanos , National Institutes of Health (U.S.) , RNA/imunologia , Pesquisa Translacional Biomédica/métodos , Estados Unidos
11.
J Extracell Vesicles ; 6(1): 1286095, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28326170

RESUMO

The release of RNA-containing extracellular vesicles (EV) into the extracellular milieu has been demonstrated in a multitude of different in vitro cell systems and in a variety of body fluids. RNA-containing EV are in the limelight for their capacity to communicate genetically encoded messages to other cells, their suitability as candidate biomarkers for diseases, and their use as therapeutic agents. Although EV-RNA has attracted enormous interest from basic researchers, clinicians, and industry, we currently have limited knowledge on which mechanisms drive and regulate RNA incorporation into EV and on how RNA-encoded messages affect signalling processes in EV-targeted cells. Moreover, EV-RNA research faces various technical challenges, such as standardisation of EV isolation methods, optimisation of methodologies to isolate and characterise minute quantities of RNA found in EV, and development of approaches to demonstrate functional transfer of EV-RNA in vivo. These topics were discussed at the 2015 EV-RNA workshop of the International Society for Extracellular Vesicles. This position paper was written by the participants of the workshop not only to give an overview of the current state of knowledge in the field, but also to clarify that our incomplete knowledge - of the nature of EV(-RNA)s and of how to effectively and reliably study them - currently prohibits the implementation of gold standards in EV-RNA research. In addition, this paper creates awareness of possibilities and limitations of currently used strategies to investigate EV-RNA and calls for caution in interpretation of the obtained data.

12.
Stem Cell Reports ; 8(3): 701-714, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28216142

RESUMO

Brain injuries, such as stroke or trauma, induce neural stem cells in the subventricular zone (SVZ) to a neurogenic response. Very little is known about the molecular cues that signal tissue damage, even over large distances, to the SVZ. Based on our analysis of gene expression patterns in the SVZ, 48 hr after an ischemic lesion caused by middle cerebral artery occlusion, we hypothesized that the presence of an injury might be transmitted by an astrocytic traveling calcium wave rather than by diffusible factors or hypoxia. Using a newly established in vitro system we show that calcium waves induced in an astrocytic monolayer spread to neural stem and progenitor cells and increase their self-renewal as well as migratory behavior. These changes are due to an upregulation of the Notch signaling pathway. This introduces the concept of propagating astrocytic calcium waves transmitting brain injury signals over long distances.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Sinalização do Cálcio , Células-Tronco Neurais/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Astrócitos/citologia , Cálcio/metabolismo , Diferenciação Celular , Movimento Celular , Autorrenovação Celular , Células Cultivadas , Modelos Animais de Doenças , Junções Comunicantes/metabolismo , Perfilação da Expressão Gênica , Masculino , Camundongos , Células-Tronco Neurais/citologia , Fatores de Tempo , Transcriptoma
14.
Oncoimmunology ; 4(6): e1008371, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26155418

RESUMO

Extracellular vesicles (EVs) have been shown to transfer various molecules, including functional RNA between cells and this process has been suggested to be particularly relevant in tumor-host interactions. However, data on EV-mediated RNA transfer has been obtained primarily by in vitro experiments or involving ex vivo manipulations likely affecting its biology, leaving their physiological relevance unclear. We engineered glioma and carcinoma tumor cells to express Cre recombinase showing their release of EVs containing Cre mRNA in various EV subfractions including exosomes. Transplantation of these genetically modified tumor cells into mice with a Cre reporter background leads to frequent recombination events at the tumor site. In both tumor models the majority of recombined cells are CD45+ leukocytes, predominantly Gr1+CD11b+ myeloid-derived suppressor cells (MDSCs). In addition, multiple lineages of recombined cells can be observed in the glioma model. In the lung carcinoma model, recombined MDSCs display an enhanced immunosuppressive phenotype and an altered miRNA profile compared to their non-recombined counterparts. Cre-lox based tracing of tumor EV RNA transfer in vivo can therefore be used to identify individual target cells in the tumor microenvironment for further mechanistical or functional analysis.

15.
PLoS Biol ; 12(6): e1001874, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24893313

RESUMO

Mechanisms behind how the immune system signals to the brain in response to systemic inflammation are not fully understood. Transgenic mice expressing Cre recombinase specifically in the hematopoietic lineage in a Cre reporter background display recombination and marker gene expression in Purkinje neurons. Here we show that reportergene expression in neurons is caused by intercellular transfer of functional Cre recombinase messenger RNA from immune cells into neurons in the absence of cell fusion. In vitro purified secreted extracellular vesicles (EVs) from blood cells contain Cre mRNA, which induces recombination in neurons when injected into the brain. Although Cre-mediated recombination events in the brain occur very rarely in healthy animals, their number increases considerably in different injury models, particularly under inflammatory conditions, and extend beyond Purkinje neurons to other neuronal populations in cortex, hippocampus, and substantia nigra. Recombined Purkinje neurons differ in their miRNA profile from their nonrecombined counterparts, indicating physiological significance. These observations reveal the existence of a previously unrecognized mechanism to communicate RNA-based signals between the hematopoietic system and various organs, including the brain, in response to inflammation.


Assuntos
Exossomos/metabolismo , Sistema Hematopoético/metabolismo , Inflamação/metabolismo , Células de Purkinje/metabolismo , RNA Mensageiro/metabolismo , Animais , Integrases , Camundongos Transgênicos , Recombinação Genética
16.
Stem Cells ; 32(1): 244-57, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24170295

RESUMO

Data from transgenic mouse models show that neuronal progenitor cells (NPCs) migrate toward experimental brain tumors and modulate the course of pathology. However, the pathways whereby NPCs are attracted to CNS neoplasms are not fully understood and it is unexplored if NPCs migrate toward brain tumors (high-grade astrocytomas) in humans. We analyzed the tumor-parenchyma interface of neurosurgical resections for the presence of (NPCs) and distinguished these physiological cells from the tumor mass. We observed that polysialic acid neural cell adhesion molecule-positive NPCs accumulate at the border of high-grade astrocytomas and display a marker profile consistent with immature migratory NPCs. Importantly, these high-grade astrocytoma-associated NPCs did not carry genetic aberrations that are indicative of the tumor. Additionally, we observed NPCs accumulating in CNS metastases. These metastatic tumors are distinguished from neural cells by defined sets of markers. Transplanting murine glioma cells embedded in a cell-impermeable hollow fiber capsule into the brains of nestin-gfp reporter mice showed that diffusible factors are sufficient to induce a neurogenic reaction. In vitro, vascular endothelial growth factor (VEGF) secreted from glioma cells increases the migratory and proliferative behavior of adult human brain-derived neural stem and progenitor cells via stimulation of VEGF receptor-2 (VEGFR-2). In vivo, inhibiting VEGFR-2 signaling with a function-blocking antibody led to a reduction in NPC migration toward tumors. Overall, our data reveal a mechanism by which NPCs are attracted to CNS tumors and suggest that NPCs accumulate in human high-grade astrocytomas.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Células-Tronco Neurais/citologia , Neurônios/citologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Movimento Celular/fisiologia , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hibridização de Ácido Nucleico
17.
Brain Pathol ; 23(1): 60-72, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22805276

RESUMO

In various types of cancers including glioblastoma, accumulating evidence show the existence of cancer stem-like cells (CSCs), characterized by stem cell marker expression, capability of differentiation and self-renewal, and high potential for tumor propagation in vivo. LGR5, whose expression is positively regulated by the Wnt signaling pathway, is a stem cell marker in intestinal mucosa and hair follicle in the skin. As Wnt signaling is also involved in brain development, the function of LGR5 in the maintenance of brain CSCs is to be assessed. Our study showed that the LGR5 transcript level was increased in CSCs. Co-immunofluorescence staining demonstrated the co-localization of CD133- and LGR5-positive cells in glioblastoma tissue sections. Functionally, silencing of LGR5 by lentiviral shRNA-mediated knockdown induced apoptosis in brain CSCs. Moreover, LGR5 depletion led to a downregulation of L1 cell adhesion molecule expression. In line with an important function in glioma tumorigenesis, LGR5 expression increased with glioma progression and correlated with an adverse outcome. Our findings suggest that LGR5 plays a role in maintenance and/or survival of brain CSCs.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patologia , Glioblastoma/diagnóstico , Glioblastoma/patologia , Células-Tronco Neoplásicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Antígeno AC133 , Antígenos CD/metabolismo , Apoptose , Diferenciação Celular/genética , Sobrevivência Celular , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/fisiologia , Glicoproteínas/metabolismo , Humanos , Lentivirus/genética , Células-Tronco Neoplásicas/patologia , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Peptídeos/metabolismo , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Estatística como Assunto , Análise Serial de Tecidos
18.
Nat Med ; 18(8): 1232-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22820645

RESUMO

Primary astrocytomas of grade 3 or 4 according to the classification system of the World Health Organization (high-grade astrocytomas or HGAs) are preponderant among adults and are almost invariably fatal despite the use of multimodal therapy. Here we show that the juvenile brain has an endogenous defense mechanism against HGAs. Neural precursor cells (NPCs) migrate to HGAs, reduce glioma expansion and prolong survival time by releasing endovanilloids that activate the vanilloid receptor (transient receptor potential vanilloid subfamily member-1 or TRPV1) on HGA cells. TRPV1 is highly expressed in tumor and weakly expressed in tumor-free brain. TRPV1 stimulation triggers tumor cell death through the branch of the endoplasmic reticulum stress pathway that is controlled by activating transcription factor-3 (ATF3). The antitumorigenic response of NPCs is lost with aging. NPC-mediated tumor suppression can be mimicked in the adult brain by systemic administration of the synthetic vanilloid arvanil, suggesting that TRPV1 agonists have potential as new HGA therapeutics.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas de Neoplasias/fisiologia , Células-Tronco Neurais/fisiologia , Canais de Cátion TRPV/fisiologia , Envelhecimento/metabolismo , Amidas , Amidoidrolases/deficiência , Amidoidrolases/genética , Animais , Antineoplásicos/uso terapêutico , Apoptose , Ácidos Araquidônicos/metabolismo , Ácidos Araquidônicos/farmacologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Movimento Celular , Meios de Cultivo Condicionados/farmacologia , Dopamina/análogos & derivados , Dopamina/metabolismo , Dopamina/farmacologia , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Etanolaminas/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Células-Tronco Neurais/metabolismo , Ácidos Oleicos/metabolismo , Ácidos Oleicos/farmacologia , Ácidos Palmíticos/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/análise , Canais de Cátion TRPV/biossíntese , Canais de Cátion TRPV/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
19.
J Virol ; 85(11): 5679-84, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21450833

RESUMO

Vesicular stomatitis virus (VSV)-based oncolytic virotherapy has the potential to significantly improve the prognosis of aggressive malignancies such as brain cancer. However, VSV's inherent neurotoxicity has hindered clinical development so far. Given that this neurotropism is attributed to the glycoprotein VSV-G, VSV was pseudotyped with the nonneurotropic envelope glycoprotein of the lymphocytic choriomeningitis virus (LCMV-GP→VSV-GP). Compared to VSV, VSV-GP showed enhanced infectivity for brain cancer cells in vitro while sparing primary human and rat neurons in vitro and in vivo, respectively. In conclusion, VSV-GP has a much wider therapeutic window than VSV and is thus more suitable for clinical applications, especially in the brain.


Assuntos
Glicoproteínas/metabolismo , Vírus da Coriomeningite Linfocítica/genética , Neuroglia/virologia , Vírus Oncolíticos/crescimento & desenvolvimento , Vesiculovirus/crescimento & desenvolvimento , Proteínas Virais/metabolismo , Tropismo Viral , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Neurônios/virologia , Vírus Oncolíticos/genética , Ratos , Infecções por Rhabdoviridae/patologia , Infecções por Rhabdoviridae/virologia , Vesiculovirus/genética
20.
Eur Heart J ; 32(5): 627-36, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21193434

RESUMO

AIMS: The identification of factors that mobilize subsets of endogenous progenitor cells may provide new therapeutic tools to enhance the repair of ischaemic tissue. We previously identified circulating mesenchymal cells that co-express endothelial markers (so-called circulating mesoangioblasts, cMABs) in children undergoing heart surgery with cardiopulmonary bypass (CPB). However, the mechanisms by which these cells are mobilized and their origin is unclear. METHODS AND RESULTS: Circulating CD73(+)CD45(-)KDR(+) cMABs were analysed in adults undergoing heart surgery with (n = 21) or without CPB (n = 8). During surgery with CPB, cMABs are mobilized with a maximal response at the end of the operation. In contrast, off-pump heart surgery does not stimulate cMAB mobilization, indicating that the stress mediated by CPB induces the mobilization of cMAB. Circulating mesoangioblasts were enriched in blood obtained from the coronary sinus. Histologically, CD73(+) cells were detected around vessels in the heart, indicating that the heart is one of the niches of cMABs. Consistently, studies in gender mismatched bone marrow transplanted patients demonstrated that cMABs did not originate from the bone marrow. Cytokine profiling of serum samples revealed that hepatocyte growth factor (HGF) was profoundly increased at the time point of maximal mobilization of cMABs. Hepatocyte growth factor stimulated the migration of cMABs. Importantly, injection of recombinant HGF increased cMABs in rats. CONCLUSIONS: Hepatocyte growth factor induces mobilization of non-haematopoietic progenitor cells with a cardiac repair capacity. This newly identified function together with the known pleiotrophic effects of HGF makes HGF an attractive therapeutic option for the treatment of ischaemic heart disease.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Leucócitos Mononucleares/citologia , Células-Tronco Mesenquimais/citologia , Idoso , Animais , Ponte Cardiopulmonar , Criança , Feminino , Humanos , Hibridização in Situ Fluorescente , Ligadura , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Infarto do Miocárdio/patologia , Ratos , Ratos Endogâmicos Lew , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...