Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
bioRxiv ; 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38352543

RESUMEN

Episodic memory involves the processing of spatial and temporal aspects of personal experiences. The lateral entorhinal cortex (LEC) plays an essential role in subserving memory. However, the specific mechanism by which LEC integrates spatial and temporal information remains elusive. Here, we recorded LEC neurons while rats performed foraging and shuttling behaviors on one-dimensional, linear or circular tracks. Unlike open-field foraging tasks, many LEC cells displayed spatial firing fields in these tasks and demonstrated selectivity for traveling directions. Furthermore, some LEC neurons displayed changes in the firing rates of their spatial rate maps during a session, a phenomenon referred to as rate remapping. Importantly, this temporal modulation was consistent across sessions, even when the spatial environment was altered. Notably, the strength of temporal modulation was found to be greater in LEC compared to other brain regions, such as the medial entorhinal cortex (MEC), CA1, and CA3. Thus, the spatial rate mapping observed in LEC neurons may serve as a coding mechanism for temporal context, allowing for flexible multiplexing of spatial and temporal information.

3.
Biochim Biophys Acta Rev Cancer ; 1879(2): 189079, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38280470

RESUMEN

Angiogenesis is a crucial process for tissue development, repair, and tumor survival. Vascular endothelial growth factor (VEGF) is a key driver secreted by cancer cells, promoting neovascularization. While VEGF's role in angiogenesis is well-documented, its influence on the other aspects in tumor microenvironemt is less discussed. This review elaborates on VEGF's impact on intercellular interactions within the tumor microenvironment, including how VEGF affects pericyte proliferation and migration and mediates interactions between tumor-associated macrophages and cancer cells, resulting in PDL-1-mediated immunosuppression and Nrf2-mediated epithelial-mesenchymal transition. The review discusses VEGF's involvement in intra-organelle crosstalk, tumor metabolism, stemness, and epithelial-mesenchymal transition. It also provides insights into current anti-VEGF therapies and their limitations in cancer treatment. Overall, this review aims to provide a thorough overview of the current state of knowledge concerning VEGF signaling and its impact, not only on angiogenesis but also on various other oncogenic processes.


Asunto(s)
60489 , Transducción de Señal , Factores de Crecimiento Endotelial Vascular , Humanos , Neoplasias/patología , Microambiente Tumoral
4.
Cancer Lett ; 578: 216455, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37865160

RESUMEN

Ubiquitin-binding associated protein 2 (UBAP2) is reported to promote macropinocytosis and pancreatic adenocarcinoma (PDAC) growth, however, its role in normal pancreatic function remains unknown. We addressed this knowledge gap by generating UBAP2 knockout (U2KO) mice under a pancreas-specific Cre recombinase (Pdx1-Cre). Pancreatic architecture remained intact in U2KO animals, but they demonstrated slight glucose intolerance compared to controls. Upon cerulein challenge to induce pancreatitis, U2KO animals had reduced levels of several pancreatitis-relevant cytokines, amylase and lipase in the serum, reduced tissue damage, and lessened neutrophil infiltration into the pancreatic tissue. Mechanistically, cerulein-challenged U2KO animals revealed reduced NF-κB activation compared to controls. In vitro promoter binding studies confirmed the reduction of NF-κB binding to its target molecules supporting UBAP2 as a new regulator of inflammation in pancreatitis and may be exploited as a therapeutic target in future to inhibit pancreatitis.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Pancreatitis , Ratones , Animales , Ceruletida/efectos adversos , FN-kappa B/metabolismo , Adenocarcinoma/patología , Neoplasias Pancreáticas/inducido químicamente , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/prevención & control , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/prevención & control , Páncreas/patología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Glucosa/metabolismo , Enfermedad Aguda
5.
ACS Nano ; 17(10): 9326-9337, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37129853

RESUMEN

The RAS-transformed cells utilize macropinocytosis to acquire amino acids to support their uncontrolled growth. However, targeting RAS to inhibit macropinocytosis remains a challenge. Here, we report that gold nanoparticles (GNP) inhibit macropinocytosis by decreasing KRAS activation. Using surface-modified and unmodified GNP, we showed that unmodified GNP specifically sequestered both wild-type and mutant KRAS and inhibited its activation, irrespective of growth factor stimulation, while surface-passivated GNP had no effect. Alteration of KRAS activation is reflected on downstream signaling cascades, macropinocytosis and tumor cell growth in vitro, and two independent preclinical human xenograft models of pancreatic cancer in vivo. The current study demonstrates NP-mediated inhibition of macropinocytosis and KRAS activation and provides translational opportunities to inhibit tumor growth in a number of cancers where activation of KRAS plays a major role.


Asunto(s)
Nanopartículas del Metal , Neoplasias Pancreáticas , Humanos , Oro/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Pinocitosis , Neoplasias Pancreáticas/patología , Proliferación Celular , Línea Celular Tumoral , Mutación
6.
Hippocampus ; 33(5): 448-464, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36965194

RESUMEN

Entorhinal cortex is the major gateway between the neocortex and the hippocampus and thus plays an essential role in subserving episodic memory and spatial navigation. It can be divided into the medial entorhinal cortex (MEC) and the lateral entorhinal cortex (LEC), which are commonly theorized to be critical for spatial (context) and non-spatial (content) inputs, respectively. Consistent with this theory, LEC neurons are found to carry little information about allocentric self-location, even in cue-rich environments, but they exhibit egocentric spatial information about external items in the environment. The superficial and deep layers of LEC are believed to mediate the input to and output from the hippocampus, respectively. As earlier studies mainly examined the spatial firing properties of superficial-layer LEC neurons, here we characterized the deep-layer LEC neurons and made direct comparisons with their superficial counterparts in single unit recordings from behaving rats. Because deep-layer LEC cells received inputs from hippocampal regions, which have strong selectivity for self-location, we hypothesized that deep-layer LEC neurons would be more informative about allocentric position than superficial-layer LEC neurons. We found that deep-layer LEC cells showed only slightly more allocentric spatial information and higher spatial consistency than superficial-layer LEC cells. Egocentric coding properties were comparable between these two subregions. In addition, LEC neurons demonstrated preferential firing at lower speeds, as well as at the boundary or corners of the environment. These results suggest that allocentric spatial outputs from the hippocampus are transformed in deep-layer LEC into the egocentric coding dimensions of LEC, rather than maintaining the allocentric spatial tuning of the CA1 place fields.


Asunto(s)
Corteza Entorrinal , Neocórtex , Ratas , Animales , Corteza Entorrinal/fisiología , Hipocampo , Neuronas/fisiología , Región CA1 Hipocampal
7.
Mater Today (Kidlington) ; 56: 79-95, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36188120

RESUMEN

The tumor microenvironment (TME) plays a key role in the poor prognosis of many cancers. However, there is a knowledge gap concerning how multicellular communication among the critical players within the TME contributes to such poor outcomes. Using epithelial ovarian cancer (EOC) as a model, we show how crosstalk among cancer cells (CC), cancer associated fibroblasts (CAF), and endothelial cells (EC) promotes EOC growth. We demonstrate here that co-culturing CC with CAF and EC promotes CC proliferation, migration, and invasion in vitro and that co-implantation of the three cell types facilitates tumor growth in vivo. We further demonstrate that disruption of this multicellular crosstalk using a gold nanoparticle (GNP) inhibits these pro-tumorigenic phenotypes in vitro as well as tumor growth in vivo. Mechanistically, GNP treatment reduces expression of several tumor-promoting cytokines and growth factors, resulting in inhibition of MAPK and PI3K-AKT activation and epithelial-mesenchymal transition - three key oncogenic signaling pathways responsible for the aggressiveness of EOC. The current work highlights the importance of multicellular crosstalk within the TME and its role for the aggressive nature of EOC, and demonstrates the disruption of these multicellular communications by self-therapeutic GNP, thus providing new avenues to interrogate the crosstalk and identify key perpetrators responsible for poor prognosis of this intractable malignancy.

8.
Adv Sci (Weinh) ; 9(31): e2200491, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36104215

RESUMEN

By exploiting the self-therapeutic properties of gold nanoparticles (GNPs) a molecular axis that promotes the growth of high-grade serous ovarian cancer (HGSOC), one of the deadliest gynecologic malignancies with poorly understood underlying molecular mechanisms, has been identified. The biodistribution and toxicity of GNPs administered by intravenous or intraperitoneal injection, both as a single dose or by repeated dosing over two weeks are first assessed; no biochemical or histological toxicity to vital organs is found. Using an orthotopic patient-derived xenograft (PDX) model of HGSOC, the authors then show that GNP treatment robustly inhibits tumor growth. Investigating the molecular mechanisms underlying the GNP efficacy reveals that GNPs downregulate insulin growth factor binding protein 2 (IGFBP2) by disrupting its autoregulation via the IGFBP2/mTOR/PTEN axis. This mechanism is validated by treating a cell line-based human xenograft tumor with GNPs and an mTOR dual-kinase inhibitor (PI-103), either individually or in combination with GNPs; GNP and PI-103 combination therapy inhibit ovarian tumor growth similarly to GNPs alone. This report illustrates how the self-therapeutic properties of GNPs can be exploited as a discovery tool to identify a critical signaling axis responsible for poor prognosis in ovarian cancer and provides an opportunity to interrogate the axis to improve patient outcomes.


Asunto(s)
Nanopartículas del Metal , Neoplasias Ováricas , Femenino , Humanos , Oro/química , Insulina , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Fosfohidrolasa PTEN , Distribución Tisular , Serina-Treonina Quinasas TOR , Animales
9.
Cancers (Basel) ; 13(5)2021 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-33802524

RESUMEN

Gynecologic malignancies, which include cancers of the cervix, ovary, uterus, vulva, vagina, and fallopian tube, are among the leading causes of female mortality worldwide, with the most prevalent being endometrial, ovarian, and cervical cancer. Gynecologic malignancies are complex, heterogeneous diseases, and despite extensive research efforts, the molecular mechanisms underlying their development and pathology remain largely unclear. Currently, mechanistic and therapeutic research in cancer is largely focused on protein targets that are encoded by about 1% of the human genome. Our current understanding of 99% of the genome, which includes noncoding RNA, is limited. The discovery of tens of thousands of noncoding RNAs (ncRNAs), possessing either structural or regulatory functions, has fundamentally altered our understanding of genetics, physiology, pathophysiology, and disease treatment as they relate to gynecologic malignancies. In recent years, it has become clear that ncRNAs are relatively stable, and can serve as biomarkers for cancer diagnosis and prognosis, as well as guide therapy choices. Here we discuss the role of small non-coding RNAs, i.e., microRNAs (miRs), P-Element induced wimpy testis interacting (PIWI) RNAs (piRNAs), and tRNA-derived small RNAs in gynecological malignancies, specifically focusing on ovarian, endometrial, and cervical cancer.

10.
Neurobiol Aging ; 98: 1-9, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33221571

RESUMEN

"Head scanning" is an investigatory behavior that has been linked to spatial exploration and the one-trial formation or strengthening of place cells in the hippocampus. Previous studies have demonstrated that a subset of aged rats with normal spatial learning performance show head scanning rates during a novel, local-global cue-mismatch manipulation that are similar to those of young rats. However, these aged rats demonstrated different patterns of expression of neural activity markers in brain regions associated with spatial learning, perhaps suggesting neural mechanisms that compensate for age-related brain changes. These prior studies did not investigate the head scanning properties of aged rats that had spatial learning impairments. The present study analyzed head scanning behavior in young, aged-unimpaired, and aged-impaired Long Evans rats. Aged-impaired rats performed the head scan behavior at a lower rate than the young rats. These results suggest that decreased attention to spatial landmarks may be a contributing factor to the spatial learning deficits shown by the aged-impaired rats.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento/psicología , Conducta Animal/fisiología , Encéfalo/fisiología , Discapacidades para el Aprendizaje/fisiopatología , Discapacidades para el Aprendizaje/psicología , Aprendizaje por Laberinto/fisiología , Aprendizaje Espacial/fisiología , Animales , Atención/fisiología , Masculino , Ratas , Ratas Long-Evans
11.
Cell Stress ; 4(11): 252-260, 2020 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-33150300

RESUMEN

Uterine carcinosarcoma (UCS) is a relatively infrequent, but extremely aggressive endometrial malignancy. Although surgery and chemotherapy have improved outcomes, overall survival (OS) remains dismal due to the lack of targeted therapy and biphasic (epithelial and mesenchymal) nature that renders the tumor aggressive and difficult to manage. Here we report a role of transforming growth factor-ß (TGFß) in maintaining epithelial to mesenchymal transition (EMT) phenotype and aggressiveness in UCS. Using a 3D-culture system, we evaluated the efficacy of the transforming growth factor-ß receptor-I (TGFßR1) kinase inhibitor Galunisertib (GLT), alone and in combination with standard chemotherapeutic drugs used for the management of UCS. We demonstrate that GLT by inhibiting canonical and non-canonical signaling emanating from transforming growth factor-ß1 (TGFß1) reduces cellular viability, invasion, clonal growth and differentiation. Interestingly, GLT sensitizes UCS cells to chemotherapy both in vitro and in in vivo preclinical tumor model. Hence, targeting TGFß signaling, in combination with standard chemotherapy, may be exploited as an important strategy to manage the clinically challenging UCS.

12.
Sci Adv ; 6(27)2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32937467

RESUMEN

The stringent expression of the hypoxia inducible factor-1α (HIF-1α) is critical to a variety of pathophysiological conditions. We reveal that, in normoxia, enzymatic action of cystathionine ß-synthase (CBS) produces H2S, which persulfidates prolyl hydroxylase 2 (PHD2) at residues Cys21 and Cys33 (zinc finger motif), augmenting prolyl hydroxylase activity. Depleting endogenous H2S either by hypoxia or by inhibiting CBS via chemical or genetic means reduces persulfidation of PHD2 and inhibits activity, preventing hydroxylation of HIF-1α, resulting in stabilization. Our in vitro findings are further supported by the depletion of CBS in the zebrafish model that exhibits axis defects and abnormal intersegmental vessels. Exogenous H2S supplementation rescues both in vitro and in vivo phenotypes. We have identified the persulfidated residues and defined their functional significance in regulating the activity of PHD2 via point mutations. Thus, the CBS/H2S/PHD2 axis may provide therapeutic opportunities for pathologies associated with HIF-1α dysregulation in chronic diseases.


Asunto(s)
Cistationina betasintasa , Subunidad alfa del Factor 1 Inducible por Hipoxia , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Animales , Cistationina betasintasa/metabolismo , Sulfuro de Hidrógeno , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Pez Cebra/metabolismo
13.
EMBO Rep ; 21(10): e48483, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32851774

RESUMEN

MICU1 is a mitochondrial inner membrane protein that inhibits mitochondrial calcium entry; elevated MICU1 expression is characteristic of many cancers, including ovarian cancer. MICU1 induces both glycolysis and chemoresistance and is associated with poor clinical outcomes. However, there are currently no available interventions to normalize aberrant MICU1 expression. Here, we demonstrate that microRNA-195-5p (miR-195) directly targets the 3' UTR of the MICU1 mRNA and represses MICU1 expression. Additionally, miR-195 is under-expressed in ovarian cancer cell lines, and restoring miR-195 expression reestablishes native MICU1 levels and the associated phenotypes. Stable expression of miR-195 in a human xenograft model of ovarian cancer significantly reduces tumor growth, increases tumor doubling times, and enhances overall survival. In conclusion, miR-195 controls MICU1 levels in ovarian cancer and could be exploited to normalize aberrant MICU1 expression, thus reversing both glycolysis and chemoresistance and consequently improving patient outcomes.


Asunto(s)
Proteínas de Transporte de Catión , MicroARNs , Neoplasias Ováricas , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Neoplasias Ováricas/genética
14.
FASEB J ; 34(9): 12024-12039, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32692445

RESUMEN

Macropinocytosis supports the metabolic requirement of RAS-transformed pancreatic ductal adenocarcinoma cells (PDACs). However, regulators of RAS-transformation (activation) that lead to macropinocytosis have not been identified. Herein, we report that UBAP2 (ubiquitin-binding associated protein 2), regulates the activation of KRAS and macropinocytosis in pancreatic cancer. We demonstrate that UBAP2 is highly expressed in both pancreatic cancer cell lines and tumor tissues of PDAC patients. The expression of UBAP2 is associated with poor overall survival in several cancers, including PDAC. Silencing UBAP2 decreases the levels of activated KRAS, and inhibits macropinocytosis, and tumor growth in vivo. Using a UBAP2-deletion construct, we demonstrate that the UBA-domain of UBAP2 is critical for the regulation of macropinocytosis and maintaining the levels of activated KRAS. In addition, UBAP2 regulates RAS downstream signaling and helps maintain RAS in the GTP-bound form. However, the exact mechanism by which UBAP2 regulates KRAS activation is unknown and needs further investigation. Thus, UBAP2 may be exploited as a potential therapeutic target to inhibit macropinocytosis and tumor growth in activated KRAS-driven cancers.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Pancreáticas/metabolismo , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Activación Enzimática , Silenciador del Gen , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Dominios Proteicos , Proteínas Proto-Oncogénicas p21(ras)/genética
15.
FASEB J ; 34(7): 9372-9392, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32463541

RESUMEN

Mutations in the human cystathionine beta synthase (CBS) gene are known to cause endothelial dysfunction responsible for cardiovascular and neurovascular diseases. CBS is the predominant hydrogen sulfide (H2 S)-producing enzyme in endothelial cells (ECs). Recently, H2 S was shown to attenuate ROS and improve mitochondrial function. Mitochondria are metabolic organelles that actively transform their ultrastructure to mediate their function. Therefore, we questioned whether perturbation of CBS/H2 S activity could drive mitochondrial dysfunction via mitochondrial dynamics in ECs. Here we demonstrate that silencing CBS induces mitochondria fragmentation, attenuates efficient oxidative phosphorylation, and decreases EC function. Mechanistically, CBS silencing significantly elevates ROS production, thereby leading to reduced mitofusin 2 (MFN2) expression, decouple endoplasmic reticulum-mitochondria contacts, increased mitochondria fission, enhanced receptor-mediated mitophagy, and increased EC death. These defects were significantly rescued by the treatment of H2 S donors. Taken together our data highlights a novel signaling axis that mechanistically links CBS with mitochondrial function and ER-mitochondrial tethering and could be considered as a new therapeutic approach for the intervention of EC dysfunction-related pathologies.


Asunto(s)
Cistationina betasintasa/metabolismo , Endotelio Vascular/fisiología , Mitocondrias/fisiología , Dinámicas Mitocondriales , Mitofagia , Estrés Oxidativo , Células Cultivadas , Retículo Endoplásmico/metabolismo , Endotelio Vascular/citología , Humanos , Transducción de Señal
16.
FASEB J ; 34(2): 2287-2300, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31908025

RESUMEN

Using a systems biology approach to prioritize potential points of intervention in ovarian cancer, we identified the lysine rich coiled-coil 1 (KRCC1), as a potential target. High-grade serous ovarian cancer patient tumors and cells express significantly higher levels of KRCC1 which correlates with poor overall survival and chemoresistance. We demonstrate that KRCC1 is predominantly present in the chromatin-bound nuclear fraction, interacts with HDAC1, HDAC2, and with the serine-threonine phosphatase PP1CC. Silencing KRCC1 inhibits cellular plasticity, invasive properties, and potentiates apoptosis resulting in reduced tumor growth. These phenotypes are associated with increased acetylation of histones and with increased phosphorylation of H2AX and CHK1, suggesting the modulation of transcription and DNA damage that may be mediated by the action of HDAC and PP1CC, respectively. Hence, we address an urgent need to develop new targets in cancer.


Asunto(s)
Daño del ADN , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Neoplasias , Neoplasias Ováricas , Transcripción Genética , Línea Celular Tumoral , Femenino , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Fosforilación , Factores de Riesgo
17.
Immunobiology ; 225(2): 151886, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31812341

RESUMEN

The objective of this study was to investigate the effect of EF24, an NF-κB-inhibitor, on the expression of negative regulators in IL-1R pathway, namely ST2 and SIGIRR. Murine JAWS II dendritic cells (DC) were challenged with lipopolysaccharide (LPS, 100 ng/ml) for 4 h, followed by treatment with 10 µM EF24 for 1 h. ST2 and SIGIRR expression was monitored by qRT-PCR and immunoblotting. ST2L and MyD88 interaction was studied by co-immunoprecipitation, and IL-33, a ST2L ligand, was assayed by ELISA. Activation of transcription factor SP1 was examined by confocal microscopy, immunoblotting, and EMSA. The effect of EF24 on accumulation of ubiquitinated proteins in DCs and proteolysis of fluorogenic peptides by purified proteasome was studied. We found that EF24 upregulated the expression of ST2 and SIGIRR and decreased the interaction of the membrane-bound ST2 (ST2L) with MyD88, and significantly reduced IL-33 levels in LPS-stimulated DCs. Simultaneously it increased the activation of transcription factor SP1and restored the basal level of ubiquitinated proteins in LPS-stimulated DCs. Moreover, EF24 inhibited trypsin- and chymotrypsin-like activity of proteasome by directly interacting with 26S proteasome. The results suggest that EF24 activates endogenous anti-inflammatory arm of IL-1R signaling, most likely by stabilizing SP1 against proteasomal degradation.


Asunto(s)
Antiinflamatorios/farmacología , Compuestos de Bencilideno/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Proteína 1 Similar al Receptor de Interleucina-1/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Piperidonas/farmacología , Receptores de Interleucina-1/antagonistas & inhibidores , Animales , Línea Celular , Interleucina-33/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Interleucina-1/metabolismo , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
ACS Appl Mater Interfaces ; 11(29): 26060-26068, 2019 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-31117437

RESUMEN

Activated cancer-associated fibroblasts (CAFs) play a major role in the poor outcome in many diseases including pancreatic cancer. Normally quiescent with high lipid content and low proliferative capacity, CAFs receiving cues from cancer cells in the tumor microenvironment become activated and transformed into a lipid-deprived and highly proliferative myofibroblast type phenotype. Therefore, reversal of activated fibroblasts to the quiescence state is an important area of investigation that may help the therapeutic management of a number of diseases including pancreatic cancer. Here, we describe a unique biological function of gold nanoparticles (GNPs) and demonstrate that GNPs may be used to transform activated CAFs to quiescence and provide insights into the underlying molecular mechanisms. Using immortalized and primary patient derived CAFs, we demonstrate that GNPs enhanced lipid content in the cells by inducing expression of lipogenesis genes such as FASN, SREBP2, and FABP3. Using pharmacological inhibitors of lipolysis, lipophagy, and fatty acid oxidation, we further demonstrate that CAFs utilized a GNP-induced endogenously synthesized lipid to maintain the quiescent phenotype. Consequently, treatment with GNP sensitizes CAF to FASN inhibitor or FASN siRNA. Hence, GNPs may be used as a tool to probe mechanisms of quiescence in CAFs and help device strategies to target the stromal compartment exploiting the mechanisms of lipid utilization.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Oro , Nanopartículas del Metal/química , Proteínas de Neoplasias/biosíntesis , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Oro/química , Oro/farmacología , Humanos , Neoplasias Pancreáticas/patología
19.
Bioconjug Chem ; 30(6): 1724-1733, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31067032

RESUMEN

It is currently recognized that perpetual cross talk among key players in tumor microenvironment such as cancer cells (CCs), cancer associated fibroblasts (CAFs), and endothelial cells (ECs) plays a critical role in tumor progression, metastasis, and therapy resistance. Disruption of the cross talk may be useful to improve the outcome of therapeutics for which limited options are available. In the current study we investigate the use of gold nanoparticles (AuNPs) as a therapeutic tool to disrupt the multicellular cross talk within the TME cells with an emphasis on inhibiting angiogenesis. We demonstrate here that AuNPs disrupt signal transduction from TME cells (CCs, CAFs, and ECs) to ECs and inhibit angiogenic phenotypes in vitro. We show that conditioned media (CM) from ovarian CCs, CAFs, or ECs themselves induce tube formation and migration of ECs in vitro. Migration of ECs is also induced when ECs are cocultured with CCs, CAFs, or ECs. In contrast, CM from the cells treated with AuNPs or cocultured cells pretreated with AuNPs demonstrate diminished effects on ECs tube formation and migration. Mechanistically, AuNPs deplete ∼95% VEGF165 from VEGF single-protein solution and remove up to ∼45% of VEGF165 from CM, which is reflected on reduced activation of VEGF-Receptor 2 (VEGFR2) as compared to control CM. These results demonstrate that AuNPs inhibit angiogenesis via blockade of VEGF-VEGFR2 signaling from TME cells to endothelial cells.


Asunto(s)
Oro/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Neovascularización Patológica/terapia , Neoplasias Ováricas/terapia , Microambiente Tumoral , Movimiento Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transducción de Señal , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Front Chem ; 6: 392, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30280096

RESUMEN

The proteasome is a validated target in drug discovery for diseases associated with unusual proteasomal activity. Here we report that two diphenyldihaloketones, CLEFMA and EF24, inhibit the peptidase activity of the 26S proteasome. The objective of this study was to investigate interaction of these compounds with the proteasome and identify a putative target within the protein components of the 26S proteasome. We employed standard fluorogenic peptide-based proteasome activity assay for trypsin-like, chymotrypsin-like, and caspase-like activities of human purified 26S proteasome in cell-free conditions. GFPu-1 and HUVEC cells were used as proteasome reporter cells. Direct binding studies used purified 19S, 20S, 26S, and recombinant RPN13-Pru for interaction with biotinylated analogs of CLEFMA and EF24. The reaction mixtures were subjected to horizontal gel electrophoresis, streptavidin-blotting, pull-down assays, and immunoblotting. The identity of the interacting protein was determined by 2D gel electrophoresis and LC-MS/MS. Drug affinity responsive target stability technique was utilized to examine if CLEFMA binding confers protection to RPN13 against thermolysin-catalyzed proteolysis. We found that trypsin-and chymotrypsin-like activities of the 26S proteasome were reduced significantly by both compounds. The compounds also reduced the proteolytic activity in GFPu-1 and HUVEC cells, resulting in accumulation of ubiquitinated proteins without affecting the autophagy process. From direct binding assays a 43 kDa protein in the 26S proteasome was found to be the interacting partner. This protein was identified by tandem mass spectroscopy as regulatory particle subunit 13 (RPN13), a ubiquitin receptor in the 19S regulatory particle. Furthermore, binding of CLEFMA to RPN13 did not protect latter from thermolysin-mediated proteolysis. Together, this study showed diphenyldihaloketones as potential proteasome inhibitors for treatment of diseases with perturbed proteasome function. The results also unraveled RPN13 as a unique target of CLEFMA and EF24. As a result, these compounds inhibit both trypsin-like and chymotrypsin-like proteasome activities.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...