Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Int J Mol Sci ; 25(2)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38256159

RESUMO

Autism spectrum disorder (ASD) is a complicated neurodevelopmental disorder, and its etiology is not well understood. It is known that genetic and nongenetic factors determine alterations in several organs, such as the liver, in individuals with this disorder. The aims of the present study were to analyze morphological and biological alterations in the liver of an autistic mouse model, BTBR T + Itpr3tf/J (BTBR) mice, and to identify therapeutic strategies for alleviating hepatic impairments using melatonin administration. We studied hepatic cytoarchitecture, oxidative stress, inflammation and ferroptosis in BTBR mice and used C57BL6/J mice as healthy control subjects. The mice were divided into four groups and then treated and not treated with melatonin, respectively. BTBR mice showed (a) a retarded development of livers and (b) iron accumulation and elevated oxidative stress and inflammation. We demonstrated that the expression of ferroptosis markers, the transcription factor nuclear factor erythroid-related factor 2 (NFR2), was upregulated, and the Kelch-like ECH-associated protein 1 (KEAP1) was downregulated in BTBR mice. Then, we evaluated the effects of melatonin on the hepatic alterations of BTBR mice; melatonin has a positive effect on liver cytoarchitecture and metabolic functions.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Ferroptose , Melatonina , Humanos , Animais , Camundongos , Transtorno Autístico/tratamento farmacológico , Transtorno Autístico/genética , Proteína 1 Associada a ECH Semelhante a Kelch , Melatonina/farmacologia , Melatonina/uso terapêutico , Transtorno do Espectro Autista/tratamento farmacológico , Transtorno do Espectro Autista/genética , Fator 2 Relacionado a NF-E2/genética , Fígado , Inflamação/tratamento farmacológico , Estresse Oxidativo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
2.
Semin Cell Dev Biol ; 92: 27-36, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30253239

RESUMO

The peritoneum is a large serous membrane with both epithelial and mesenchymal features, and is essential for maintaining an intra-abdominal homeostatic equilibrium. The peritoneum plays a central role in the pathogenesis of a number of disorders. Pathological processes affecting the peritoneum such as inflammation and carcinomatosis can have serious clinical consequences, but the pathophysiology of these conditions is poorly understood. Understanding peritoneal embryology, anatomy and physiology is crucial to comprehend pathophysiological mechanisms and to devise a new focus for research. The vascular response to pathological processes appears to be of considerable importance, since the peritoneal vasculature plays a pivotal role in most associated diseases. Therefore, this review summarizes currently available literature with special emphasis on the development, anatomy and function of the peritoneal vasculature. Pathological processes are described to illustrate physiological and pathophysiological characteristics of the peritoneum.


Assuntos
Peritônio , Humanos , Peritônio/anatomia & histologia , Peritônio/embriologia , Peritônio/fisiologia , Peritônio/fisiopatologia
3.
Biochim Biophys Acta Rev Cancer ; 1869(2): 346-354, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29684521

RESUMO

Glioblastoma is the most lethal primary brain tumor and poor survival of glioblastoma patients is attributed to the presence of glioma stem cells (GSCs). These therapy-resistant, quiescent and pluripotent cells reside in GSC niches, which are specific microenvironments that protect GSCs against radiotherapy and chemotherapy. We previously showed the existence of hypoxic peri-arteriolar GSC niches in glioblastoma tumor samples. However, other studies have described peri-vascular niches, peri-hypoxic niches, peri-immune niches and extracellular matrix niches of GSCs. The aim of this review was to critically evaluate the literature on these five different types of GSC niches. In the present review, we describe that the five niche types are not distinct from one another, but should be considered to be parts of one integral GSC niche model, the hypoxic peri-arteriolar GSC niche. Moreover, hypoxic peri-arteriolar GSC niches are structural and functional look-alikes of hematopoietic stem cell (HSC) niches in the bone marrow. GSCs are maintained in peri-arteriolar niches by the same receptor-ligand interactions as HSCs in bone marrow. Our concept should be rigidly tested in the near future and applied to develop therapies to expel and keep GSCs out of their protective niches to render them more vulnerable to standard therapies.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Oxigênio/metabolismo , Nicho de Células-Tronco , Hipóxia Tumoral , Microambiente Tumoral , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação
4.
FASEB J ; 33(4): 5320-5333, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30698992

RESUMO

Insight into the molecular and cellular processes in blood-retinal barrier (BRB) development, including the contribution of paracellular and transcellular pathways, is still incomplete but may help to understand the inverse process of BRB loss in pathologic eye conditions. In this comprehensive observational study, we describe in detail the formation of the BRB at the molecular level in physiologic conditions, using mice from postnatal day (P)3 to P25. Our data indicate that immature blood vessels already have tight junctions at P5, before the formation of a functional BRB. Expression of the endothelial cell-specific protein plasmalemma vesicle-associated protein (PLVAP), which is known to be involved in transcellular transport and associated with BRB permeability, decreased during development and was absent when a functional barrier was formed. Moreover, we show that PLVAP deficiency causes a transient delay in retinal vascular development and changes in mRNA expression levels of endothelial permeability pathway proteins.-Van der Wijk, A.-E., Wisniewska-Kruk, J., Vogels, I. M. C., van Veen, H. A., Ip, W. F., van der Wel, N. N., van Noorden, C. J. F., Schlingemann, R. O., Klaassen, I. Expression patterns of endothelial permeability pathways in the development of the blood-retinal barrier in mice.


Assuntos
Barreira Hematorretiniana/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/genética , Animais , Barreira Hematorretiniana/embriologia , Barreira Hematorretiniana/ultraestrutura , Western Blotting , Éxons/genética , Genótipo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma
5.
FASEB J ; 33(1): 557-571, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30001166

RESUMO

Diffuse gliomas often carry point mutations in isocitrate dehydrogenase ( IDH1mut), resulting in metabolic stress. Although IDHmut gliomas are difficult to culture in vitro, they thrive in the brain via diffuse infiltration, suggesting brain-specific tumor-stroma interactions that can compensate for IDH-1 deficits. To elucidate the metabolic adjustments in clinical IDHmut gliomas that contribute to their malignancy, we applied a recently developed method of targeted quantitative RNA next-generation sequencing to 66 clinical gliomas and relevant orthotopic glioma xenografts, with and without the endogenous IDH-1R132H mutation. Datasets were analyzed in R using Manhattan plots to calculate distance between expression profiles, Ward's method to perform unsupervised agglomerative clustering, and the Mann Whitney U test and Fisher's exact tests for supervised group analyses. The significance of transcriptome data was investigated by protein analysis, in situ enzymatic activity mapping, and in vivo magnetic resonance spectroscopy of orthotopic IDH1mut- and IDHwt-glioma xenografts. Gene set enrichment analyses of clinical IDH1mut gliomas strongly suggest a role for catabolism of lactate and the neurotransmitter glutamate, whereas, in IDHwt gliomas, processing of glucose and glutamine are the predominant metabolic pathways. Further evidence of the differential metabolic activity in these cancers comes from in situ enzymatic mapping studies and preclinical in vivo magnetic resonance spectroscopy imaging. Our data support an evolutionary model in which IDHmut glioma cells exist in symbiosis with supportive neuronal cells and astrocytes as suppliers of glutamate and lactate, possibly explaining the diffuse nature of these cancers. The dependency on glutamate and lactate opens the way for novel approaches in the treatment of IDHmut gliomas.-Lenting, K., Khurshed, M., Peeters, T. H., van den Heuvel, C. N. A. M., van Lith, S. A. M., de Bitter, T., Hendriks, W., Span, P. N., Molenaar, R. J., Botman, D., Verrijp, K., Heerschap, A., ter Laan, M., Kusters, B., van Ewijk, A., Huynen, M. A., van Noorden, C. J. F., Leenders, W. P. J. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Ácido Glutâmico/metabolismo , Isocitrato Desidrogenase/genética , Ácido Láctico/metabolismo , Mutação , Estresse Fisiológico , 4-Aminobutirato Transaminase/genética , 4-Aminobutirato Transaminase/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/metabolismo , Glutamato Desidrogenase/genética , Glutamato Desidrogenase/metabolismo , Glutaminase/genética , Glutaminase/metabolismo , Humanos , Isocitrato Desidrogenase/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Succinato-Semialdeído Desidrogenase/genética , Succinato-Semialdeído Desidrogenase/metabolismo , Transcriptoma , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Biochim Biophys Acta Rev Cancer ; 1868(1): 183-198, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28363872

RESUMO

Acute myeloid leukemia and acute lymphoblastic leukemia cells hijack hematopoietic stem cell (HSC) niches in the bone marrow and become leukemic stem cells (LSCs) at the expense of normal HSCs. LSCs are quiescent and resistant to chemotherapy and can cause relapse of the disease. HSCs in niches are needed to generate blood cell precursors that are committed to unilineage differentiation and eventually production of mature blood cells, including red blood cells, megakaryocytes, myeloid cells and lymphocytes. Thus far, three types of HSC niches are recognized: endosteal, reticular and perivascular niches. However, we argue here that there is only one type of HSC niche, which consists of a periarteriolar compartment and a perisinusoidal compartment. In the periarteriolar compartment, hypoxia and low levels of reactive oxygen species preserve the HSC pool. In the perisinusoidal compartment, hypoxia in combination with higher levels of reactive oxygen species enables proliferation of progenitor cells and their mobilization into the circulation. Because HSC niches offer protection to LSCs against chemotherapy, we review novel therapeutic strategies to inhibit homing of LSCs in niches for the prevention of dedifferentiation of leukemic cells into LSCs and to stimulate migration of leukemic cells out of niches. These strategies enhance differentiation and proliferation and thus sensitize leukemic cells to chemotherapy. Finally, we list clinical trials of therapies that tackle LSCs in HSC niches to circumvent their protection against chemotherapy.


Assuntos
Leucemia Mieloide Aguda/terapia , Células-Tronco Neoplásicas/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos
7.
Mol Biol Rep ; 47(4): 2561-2572, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32133604

RESUMO

PURPOSE: We have previously identified insulin-like growth factor 2 (IGF2) and insulin-like growth factor 1 receptor (IGF1R) as essential proteins for tip cell maintenance and sprouting angiogenesis. In this study, we aim to identify other IGF family members involved in endothelial sprouting angiogenesis. METHODS: Effects on sprouting were analyzed in human umbilical vein endothelial cells (HUVECs) using the spheroid-based sprouting model, and were quantified as mean number of sprouts per spheroid and average sprout length. RNA silencing technology was used to knockdown gene expression. Recombinant forms of the ligands (IGF1 and IGF2, insulin) and the IGF-binding proteins (IGFBP) 3 and 4 were used to induce excess effects. Effects on the tip cell phenotype were analyzed by measuring the fraction of CD34+ tip cells using flow cytometry and immunohistochemistry in a 3D angiogenesis model. Experiments were performed in the presence and absence of serum. RESULTS: Knockdown of IGF2 inhibited sprouting in HUVECs, in particular when cultured in the absence of serum, suggesting that components in serum influence the signaling of IGF2 in angiogenesis in vitro. We then determined the effects of IGFBP3 and IGFBP4, which are both present in serum, on IGF2-IGF1R signaling in sprouting angiogenesis in the absence of serum: knockdown of IGFBP3 significantly reduced sprouting angiogenesis, whereas knockdown of IGFBP4 resulted in increased sprouting angiogenesis in both flow cytometry analysis and immunohistochemical analysis of the 3D angiogenesis model. Other IGF family members except INSR did not affect IGF2-IGF1R signaling. CONCLUSIONS: Serum components and IGF binding proteins regulate IGF2 effects on sprouting angiogenesis. Whereas IGFBP3 acts as co-factor for IGF2-IGF1R binding, IGFBP4 inhibits IGF2 signaling.


Assuntos
Indutores da Angiogênese/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Fator de Crescimento Insulin-Like I , Fator de Crescimento Insulin-Like II , Morfogênese , Neovascularização Patológica/metabolismo , Organoides/metabolismo , Receptor IGF Tipo 1 , Receptor IGF Tipo 2 , Transdução de Sinais
8.
Curr Treat Options Oncol ; 21(2): 17, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32025928

RESUMO

OPINION STATEMENT: Cancer-related fatigue (CRF) is a problem for a significant proportion of cancer survivors during and after active cancer treatment. However, CRF is underdiagnosed and undertreated. Interventions are available for CRF although there is no gold standard. Based on current level of evidence, exercise seems to be most effective in preventing or ameliorating CRF during the active- and posttreatment phases.


Assuntos
Fadiga/etiologia , Fadiga/terapia , Neoplasias/complicações , Fatores Etários , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Sobreviventes de Câncer , Tomada de Decisão Clínica , Terapia Combinada , Comorbidade , Gerenciamento Clínico , Suscetibilidade a Doenças , Fadiga/diagnóstico , Fadiga/epidemiologia , Humanos , Neoplasias/diagnóstico , Neoplasias/epidemiologia , Prevalência , Radioterapia/efeitos adversos , Radioterapia/métodos , Fatores de Risco , Sobrevivência , Resultado do Tratamento
9.
Acta Obstet Gynecol Scand ; 99(8): 983-993, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32108320

RESUMO

Low-lying placentas, placenta previa and abnormally invasive placentas are the most frequently occurring placental abnormalities in location and anatomy. These conditions can have serious consequences for mother and fetus mainly due to excessive blood loss before, during or after delivery. The incidence of such abnormalities is increasing, but treatment options and preventive strategies are limited. Therefore, it is crucial to understand the etiology of placental abnormalities in location and anatomy. Placental formation already starts at implantation and therefore disorders during implantation may cause these abnormalities. Understanding of the normal placental structure and development is essential to comprehend the etiology of placental abnormalities in location and anatomy, to diagnose the affected women and to guide future research for treatment and preventive strategies. We reviewed the literature on the structure and development of the normal placenta and the placental development resulting in low-lying placentas, placenta previa and abnormally invasive placentas.


Assuntos
Doenças Placentárias/fisiopatologia , Doenças Placentárias/terapia , Adulto , Feminino , Humanos , Gravidez
10.
FASEB J ; : fj201800547R, 2018 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-29879375

RESUMO

Isocitrate dehydrogenase ( IDH1)-1 is mutated in various types of human cancer, and the presence of this mutation is associated with improved responses to irradiation and chemotherapy in solid tumor cells. Mutated IDH1 (IDH1MUT) enzymes consume NADPH to produce d-2-hydroxyglutarate (d-2HG) resulting in the decreased reducing power needed for detoxification of reactive oxygen species (ROS), for example. The objective of the current study was to investigate the mechanism behind the chemosensitivity of the widely used anticancer agent cisplatin in IDH1MUT cancer cells. Oxidative stress, DNA damage, and mitochondrial dysfunction caused by cisplatin treatment were monitored in IDH1MUT HCT116 colorectal cancer cells and U251 glioma cells. We found that exposure to cisplatin induced higher levels of ROS, DNA double-strand breaks (DSBs), and cell death in IDH1MUT cancer cells, as compared with IDH1 wild-type ( IDH1WT) cells. Mechanistic investigations revealed that cisplatin treatment dose dependently reduced oxidative respiration in IDH1MUT cells, which was accompanied by disturbed mitochondrial proteostasis, indicative of impaired mitochondrial activity. These effects were abolished by the IDH1MUT inhibitor AGI-5198 and were restored by treatment with d-2HG. Thus, our study shows that altered oxidative stress responses and a vulnerable oxidative metabolism underlie the sensitivity of IDH1MUT cancer cells to cisplatin.-Khurshed, M., Aarnoudse, N., Hulsbos, R., Hira, V. V. V., van Laarhoven, H. W. M., Wilmink, J. W., Molenaar, R. J., van Noorden, C. J. F. IDH1-mutant cancer cells are sensitive to cisplatin and an IDH1-mutant inhibitor counteracts this sensitivity.

11.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 594-603, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28040478

RESUMO

Glioblastoma (GBM) is the most aggressive primary brain tumor with poor patient survival that is at least partly caused by malignant and therapy-resistant glioma stem-like cells (GSLCs) that are protected in GSLC niches. Previously, we have shown that the chemo-attractant stromal-derived factor-1α (SDF-1α), its C-X-C receptor type 4 (CXCR4) and the cysteine protease cathepsin K (CatK) are localized in GSLC niches in glioblastoma. Here, we investigated whether SDF-1α is a niche factor that through its interactions with CXCR4 and/or its second receptor CXCR7 on GSLCs facilitates their homing to niches. Furthermore, we aimed to prove that SDF-1α cleavage by CatK inactivates SDF-1α and inhibits the invasion of GSLCs. We performed mass spectrometric analysis of cleavage products of SDF-1α after proteolysis by CatK. We demonstrated that CatK cleaves SDF-1α at 3 sites in the N-terminus, which is the region of SDF-1α that binds to its receptors. Confocal imaging of human GBM tissue sections confirmed co-localization of SDF-1α and CatK in GSLC niches. In accordance, 2D and 3D invasion experiments using CXCR4/CXCR7-expressing GSLCs and GBM cells showed that SDF-1α had chemotactic activity whereas CatK cleavage products of SDF-1α did not. Besides, CXCR4 inhibitor plerixafor inhibited invasion of CXCR4/CXCR7-expressing GSLCs. In conclusion, CatK can cleave and inactivate SDF-1α. This implies that CatK activity facilitates migration of GSLCs out of niches. We propose that activation of CatK may be a promising strategy to prevent homing of GSLCs in niches and thus render these cells sensitive to chemotherapy and radiation.


Assuntos
Catepsina K/metabolismo , Quimiocina CXCL12/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neuroglia/metabolismo , Receptores CXCR4/metabolismo , Sequência de Aminoácidos , Benzilaminas , Catepsina K/genética , Linhagem Celular Tumoral , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiotaxia , Ciclamos , Expressão Gênica , Compostos Heterocíclicos/farmacologia , Humanos , Células-Tronco Neoplásicas/patologia , Neuroglia/patologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteólise , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Nicho de Células-Tronco/genética
12.
Angiogenesis ; 21(4): 823-836, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29951828

RESUMO

Tip cells, the leading cells of angiogenic sprouts, were identified in cultures of human umbilical vein endothelial cells (HUVECs) by using CD34 as a marker. Here, we show that tip cells are also present in primary human microvascular endothelial cells (hMVECs), a more relevant endothelial cell type for angiogenesis. By means of flow cytometry, immunocytochemistry, and qPCR, it is shown that endothelial cell cultures contain a dynamic population of CD34+ cells with many hallmarks of tip cells, including filopodia-like extensions, elevated mRNA levels of known tip cell genes, and responsiveness to stimulation with VEGF and inhibition by DLL4. Furthermore, we demonstrate that our in vitro tip cell model can be exploited to investigate cellular and molecular mechanisms in tip cells and to discover novel targets for anti-angiogenesis therapy in patients. Small interfering RNA (siRNA) was used to knockdown gene expression of the known tip cell genes angiopoietin 2 (ANGPT2) and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), which resulted in similar effects on tip cells and sprouting as compared to inhibition of tip cells in vivo. Finally, we identified two novel tip cell-specific genes in CD34+ tip cells in vitro: insulin-like growth factor 2 (IGF2) and IGF-1-receptor (IGF1R). Knockdown of these genes resulted in a significant decrease in the fraction of tip cells and in the extent of sprouting in vitro and in vivo. In conclusion, this study shows that by using our in vitro tip cell model, two novel essential tip cells genes are identified.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Microvasos/metabolismo , Receptores de Somatomedina/metabolismo , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Embrião de Galinha , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Fator de Crescimento Insulin-Like II/genética , Microvasos/citologia , Receptor IGF Tipo 1 , Receptor de TIE-1/genética , Receptor de TIE-1/metabolismo , Receptores de Somatomedina/genética , Peixe-Zebra
13.
Nutr Cancer ; 70(3): 317-333, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29570984

RESUMO

The review discusses the effects of Epigallocatechin-3-gallate Gallate (EGCG) on glioma as a basis for future research on clinical application of EGCG. Epidemiological studies on the effects of green tea or EGCG on the risk of glioma is inconclusive due to the limited number of studies, the inclusion of all tea types in these studies, and the focus on caffeine rather than EGCG. In vivo experiments using EGCG monotherapy are inconclusive. Nevertheless, EGCG induces cell death, prevents cellular proliferation, and limits invasion in multiple glioma cell lines. Furthermore, EGCG enhances the efficacy of anti-glioma therapies, including irradiation, temozolomide, carmustine, cisplatin, tamoxifen, and TNF-related apoptosis-inducing ligand, but reduces the effect of bortezomib. Pro-drugs, co-treatment, and encapsulation are being investigated to enhance clinical applicability of EGCG. Mechanisms of actions of EGCG have been partly elucidated. EGCG has both anti-oxidant and oxidant properties. EGCG inhibits pro-survival proteins, such as telomerase, survivin, GRP78, PEA15, and P-gp. EGCG inhibits signaling of PDGFR, IGF-1R, and 67LR. EGCG reduces invasiveness of cancer cells by inhibiting the activities of various metalloproteinases, cytokines, and chemokines. Last, EGCG inhibits some NADPH-producing enzymes, thus disturbing redox status and metabolism of glioma cells. In conclusion, EGCG may be a suitable adjuvant to potentiate anti-glioma therapies.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Catequina/análogos & derivados , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Glioma/tratamento farmacológico , Chá/química , Animais , Anticarcinógenos/farmacologia , Antineoplásicos Fitogênicos/farmacocinética , Catequina/farmacocinética , Catequina/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Central/terapia , Terapia Combinada , Chaperona BiP do Retículo Endoplasmático , Estudos Epidemiológicos , Glioma/patologia , Glioma/terapia , Humanos , Neoplasias Experimentais/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos
14.
Ophthalmic Res ; 59(2): 98-109, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29073609

RESUMO

INTRODUCTION: Curcumin has multiple biological effects including the modulation of protein homeostasis by the ubiquitin-proteasome system. The purpose of this study was to assess the in vitro cytotoxic and oxidative effects of nano-curcumin and standard curcumin and characterize their effects on proteasome regulation in retinal pigment epithelial (RPE) cells. METHODS: Viability, cell cycle progression, and reactive oxygen species (ROS) production were determined after treatment with nano-curcumin or curcumin. Subsequently, the effects of nano-curcumin and curcumin on proteasome activity and the gene and protein expression of proteasome subunits PA28α, α7, ß5, and ß5i were assessed. RESULTS: Nano-curcumin (5-100 µM) did not show significant cytotoxicity or anti-oxidative effects against H2O2-induced oxidative stress, whereas curcumin (≥10 µM) was cytotoxic and a potent inducer of ROS production. Both nano-curcumin and curcumin induced changes in proteasome-mediated proteolytic activity characterized by increased activity of the proteasome subunits ß2 and ß5i/ß1 and reduced activity of ß5/ß1i. Likewise, nano-curcumin and curcumin affected mRNA and protein levels of household and immunoproteasome subunits. CONCLUSIONS: Nano-curcumin is less toxic to RPE cells and less prone to induce ROS production than curcumin. Both nano-curcumin and curcumin increase proteasome-mediated proteolytic activity. These results suggest that nano-curcumin may be regarded as a proteasome-modulating agent of limited cytotoxicity for RPE cells.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Curcumina/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Pigmentos da Retina/metabolismo
15.
Lasers Med Sci ; 33(2): 407-423, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29177555

RESUMO

Meta-analysis of treatment effects of antimicrobial photodynamic therapy (aPDT) adjunct to non-surgical scaling and root planing (SRP) in comparison to SRP alone on patients with chronic periodontitis. The meta-analysis was performed according to PRISMA statement and Cochrane Collaboration guidelines. Electronic search complemented by hand search assured a high yield of randomized controlled trials (RCTs) of aPDT as adjunct modality to SRP. Differences in probing depth (PD) and clinical attachment level (CAL) were calculated with 95% confidence intervals and pooled in a random effects model. Analysis for intra- and inter-study heterogeneity was provided by χ 2 and I 2 tests, and publication bias was checked by funnel plots. Pooled overall effects of 26 RCTs attested significant benefits of aPDT adjunct to SRP with respect to PD reduction (MD 0.37; 95% CI 0.12-0.53; P < 0.0001) and CAL gain (MD 0.33; 95% CI 0.19-0.48; P < 0.00001) after 3 and 6 months. Sensitivity analysis minimized heterogeneity of PD reduction (MD 0.21; 95% CI 0.13-0.30; P < 0.00001) and CAL gain (MD 0.36; 95% CI 0.27-0.46). aPDT adjunct to SRP provides significant PD reduction and CAL gain in treatment of chronic periodontitis. This moderate effect was found after 3 and 6 months which is short from a clinical perspective.


Assuntos
Periodontite Crônica/tratamento farmacológico , Fotoquimioterapia , Feminino , Seguimentos , Humanos , Masculino , Viés de Publicação , Resultado do Tratamento
16.
Am J Physiol Renal Physiol ; 312(3): F457-F464, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031169

RESUMO

The hypoxanthine-xanthine oxidase (XO) axis is considered to be a key driver of transplantation-related ischemia-reperfusion (I/R) injury. Whereas interference with this axis effectively quenches I/R injury in preclinical models, there is limited efficacy of XO inhibitors in clinical trials. In this context, we considered clinical evaluation of a role for the hypoxanthine-XO axis in human I/R to be relevant. Patients undergoing renal allograft transplantation were included (n = 40) and classified based on duration of ischemia (short, intermediate, and prolonged). Purine metabolites excreted by the reperfused kidney (arteriovenous differences) were analyzed by the ultra performance liquid chromatography-tandem mass spectrometer (UPLCMS/MS) method and tissue XO activity was assessed by in situ enzymography. We confirmed progressive hypoxanthine accumulation (P < 0.006) during ischemia, using kidney transplantation as a clinical model of I/R. Yet, arteriovenous concentration differences of uric acid and in situ enzymography of XO did not indicate significant XO activity in ischemic and reperfused kidney grafts. Furthermore, we tested a putative association between hypoxanthine accumulation and renal oxidative stress by assessing renal malondialdehyde and isoprostane levels and allantoin formation during the reperfusion period. Absent release of these markers is not consistent with an association between ischemic hypoxanthine accumulation and postreperfusion oxidative stress. On basis of these data for the human kidney we hypothesize that the role for the hypoxanthine-XO axis in clinical I/R injury is less than commonly thought, and as such the data provide an explanation for the apparent limited clinical efficacy of XO inhibitors.


Assuntos
Função Retardada do Enxerto/enzimologia , Hipoxantina/metabolismo , Transplante de Rim/efeitos adversos , Rim/enzimologia , Rim/cirurgia , Traumatismo por Reperfusão/enzimologia , Xantina Oxidase/metabolismo , Adulto , Idoso , Biomarcadores/sangue , Cromatografia Líquida de Alta Pressão , Função Retardada do Enxerto/diagnóstico , Função Retardada do Enxerto/etiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Espécies Reativas de Oxigênio/sangue , Traumatismo por Reperfusão/diagnóstico , Traumatismo por Reperfusão/etiologia , Transdução de Sinais , Espectrometria de Massas em Tandem , Fatores de Tempo , Resultado do Tratamento , Ácido Úrico/sangue , Xantina Oxidase/sangue
17.
Am J Pathol ; 186(4): 1044-54, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26878208

RESUMO

Loss of blood-retinal barrier (BRB) properties induced by vascular endothelial growth factor (VEGF) and other factors is an important cause of diabetic macular edema. Previously, we found that the presence of plasmalemma vesicle-associated protein (PLVAP) in retinal capillaries associates with loss of BRB properties and correlates with increased vascular permeability in diabetic macular edema. In this study, we investigated whether absence of PLVAP protects the BRB from VEGF-induced permeability. We used lentiviral-delivered shRNA or siRNA to inhibit PLVAP expression. The barrier properties of in vitro BRB models were assessed by measuring transendothelial electrical resistance, permeability of differently sized tracers, and the presence of endothelial junction complexes. The effect of VEGF on caveolae formation was studied in human retinal explants. BRB loss in vivo was studied in the mouse oxygen-induced retinopathy model. The inhibition of PLVAP expression resulted in decreased VEGF-induced BRB permeability of fluorescent tracers, both in vivo and in vitro. PLVAP inhibition attenuated transendothelial electrical resistance reduction induced by VEGF in BRB models in vitro and significantly increased transendothelial electrical resistance of the nonbarrier human umbilical vein endothelial cells. Furthermore, PLVAP knockdown prevented VEGF-induced caveolae formation in retinal explants but did not rescue VEGF-induced alterations in endothelial junction complexes. In conclusion, PLVAP is an essential cofactor in VEGF-induced BRB permeability and may become an interesting novel target for diabetic macular edema therapy.


Assuntos
Barreira Hematorretiniana/metabolismo , Permeabilidade Capilar/fisiologia , Retinopatia Diabética/metabolismo , Retina/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/ultraestrutura , Animais , Permeabilidade Capilar/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Edema Macular/metabolismo , Edema Macular/patologia , Camundongos , Ratos , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Semin Cancer Biol ; 35: 71-84, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26320409

RESUMO

Proteases, including lysosomal cathepsins, are functionally involved in many processes in cancer progression from its initiation to invasion and metastatic spread. Only recently, cathepsin K (CatK), the cysteine protease originally reported as a collagenolytic protease produced by osteoclasts, appeared to be overexpressed as well in various types of cancers. In this review, the physiological functions of CatK are presented and compared to its potential role in pathobiolology of processes associated with tumour growth, invasion and metastasis of cancer cells and their interactions with the tumour microenvironment. CatK activity is either indirectly affecting signalling pathways, or directly degrading extracellular matrix (ECM) proteins, for example in bone metastases. Recently, CatK was also found in glioma, possibly regulating cancer stem-like cell mobilisation and modulating recently found physiological CatK substrates, including chemokines and growth factors. Moreover, CatK may be useful in differential diagnosis and may have prognostic value. Finally, the application of CatK inhibitors, which are already in clinical trials for treatment of osteoporosis, has a potential to attenuate cancer aggressiveness.


Assuntos
Catepsina K/metabolismo , Neoplasias/metabolismo , Peptídeo Hidrolases/metabolismo , Animais , Biomarcadores Tumorais , Catepsina K/antagonistas & inibidores , Catepsina K/genética , Progressão da Doença , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Lisossomos/enzimologia , Lisossomos/metabolismo , Terapia de Alvo Molecular , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Peptídeo Hidrolases/genética , Prognóstico , Proteólise , Resultado do Tratamento , Microambiente Tumoral/genética
20.
Transfusion ; 56(2): 427-32, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26456480

RESUMO

BACKGROUND: During storage, the activity of the red blood cell (RBC) antioxidant system decreases. Glucose-6-phosphate dehydrogenase (G6PD) is essential for protection against oxidative stress by producing NADPH. G6PD function of RBC transfusion products is reported to remain stable during storage, but activity was measured in hemolysates and not in individual RBCs. We hypothesized that analysis of G6PD activity in individual RBC identifies storage-dependent changes in G6PD function. STUDY DESIGN AND METHODS: Seven units of stored leukoreduced RBCs, stored in saline-adenine-glucose-mannitol, were sampled every week up to 6 weeks of storage. G6PD activity was determined with the cytofluorometric method and expressed as mean fluorescent intensity (MFI) per RBC. RESULTS: During storage, G6PD activity decreased significantly. Mean MFI after 3 days of storage was 27.8 ± 8.8 and gradually decreased significantly to 18.0 ± 8.3 after 42 days. CONCLUSION: G6PD activity decreases during storage of leukoreduced RBCs. Our results may form a new target to improve storage conditions of RBCs and subsequently improve the quality of transfusion products.


Assuntos
Preservação de Sangue , Eritrócitos/enzimologia , Glucosefosfato Desidrogenase/metabolismo , Eritrócitos/citologia , Glucosefosfato Desidrogenase/química , Humanos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa