Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Adv ; 9(23): eadg3988, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37285426

RESUMEN

Implanted electronic sensors, compared with conventional medical imaging, allow monitoring of advanced physiological properties of soft biological tissues continuously, such as adhesion, pH, viscoelasticity, and biomarkers for disease diagnosis. However, they are typically invasive, requiring being deployed by surgery, and frequently cause inflammation. Here we propose a minimally invasive method of using wireless miniature soft robots to in situ sense the physiological properties of tissues. By controlling robot-tissue interaction using external magnetic fields, visualized by medical imaging, we can recover tissue properties precisely from the robot shape and magnetic fields. We demonstrate that the robot can traverse tissues with multimodal locomotion and sense the adhesion, pH, and viscoelasticity on porcine and mice gastrointestinal tissues ex vivo, tracked by x-ray or ultrasound imaging. With the unprecedented capability of sensing tissue physiological properties with minimal invasion and high resolution deep inside our body, this technology can potentially enable critical applications in both basic research and clinical practice.


Asunto(s)
Robótica , Porcinos , Animales , Ratones , Locomoción , Tecnología , Diseño de Equipo
2.
Am J Physiol Gastrointest Liver Physiol ; 321(6): G693-G704, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34643092

RESUMEN

All-time preservation of an intact mucosal barrier is crucial to ensuring intestinal homeostasis and, hence, the organism's overall health maintenance. This complex process relies on an equilibrated signaling system between the intestinal epithelium and numerous cell populations inhabiting the gut mucosa. Any perturbations of this delicate cross talk, particularly regarding the immune cell compartment and microbiota, may sustainably debilitate the intestinal barrier function. As a final joint event, a critical rise in epithelial permeability facilitates the exposure of submucosal immunity to microbial antigens, resulting in uncontrolled inflammation, collateral tissue destruction, and dysbiosis. Organoid-derived intestinal coculture models have established themselves as convenient tools to reenact such pathophysiological events, explore interactions between selected cell populations, and assess their roles with a central focus on intestinal barrier recovery and stabilization.


Asunto(s)
Mucosa Intestinal/citología , Organoides/citología , Cultivo Primario de Células/métodos , Animales , Técnicas de Cocultivo/métodos , Microbioma Gastrointestinal , Humanos , Mucosa Intestinal/microbiología , Linfocitos/citología , Macrófagos/citología , Organoides/microbiología
3.
Front Immunol ; 11: 2081, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983160

RESUMEN

Trauma represents a major socioeconomic burden worldwide. After a severe injury, hemorrhagic shock (HS) as a frequent concomitant aspect is a central driver of systemic inflammation and organ damage. The kidney is often strongly affected by traumatic-HS, and acute kidney injury (AKI) poses the patient at great risk for adverse outcome. Recently, thirty-eight-negative kinase 1 (TNK1) was proposed to play a detrimental role in organ damage after trauma/HS. Therefore, we aimed to assess the role of TNK1 in HS-induced kidney injury in a murine and a post hoc analysis of a non-human primate model of HS comparable to the clinical situation. Mice and non-human primates underwent resuscitated HS at 30 mmHg for 60 min. 5 h after the induction of shock, animals were assessed for systemic inflammation and TNK1 expression in the kidney. In vitro, murine distal convoluted tubule cells were stimulated with inflammatory mediators to gain mechanistic insights into the role of TNK1 in kidney dysfunction. In a translational approach, we investigated blood drawn from either healthy volunteers or severely injured patients at different time points after trauma (from arrival at the emergency room and at fixed time intervals until 10 days post injury; identifier: NCT02682550, https://clinicaltrials.gov/ct2/show/NCT02682550). A pronounced inflammatory response, as seen by increased IL-6 plasma levels as well as early signs of AKI, were observed in mice, non-human primates, and humans after trauma/HS. TNK1 was found in the plasma early after trauma-HS in trauma patients. Renal TNK1 expression was significantly increased in mice and non-human primates after HS, and these effects with concomitant induction of apoptosis were blocked by therapeutic inhibition of complement C3 activation in non-human primates. Mechanistically, in vitro data suggested that IL-6 rather than C3 cleavage products induced upregulation of TNK1 and impaired barrier function in renal epithelial cells. In conclusion, these data indicate that C3 inhibition in vivo may inhibit an excessive inflammatory response and mediator release, thereby indirectly neutralizing TNK1 as a potent driver of organ damage. In future studies, we will address the therapeutic potential of direct TNK1 inhibition in the context of severe tissue trauma with different degrees of additional HS.


Asunto(s)
Proteínas Fetales/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Choque Hemorrágico/metabolismo , Heridas y Lesiones/metabolismo , Lesión Renal Aguda , Animales , Células Cultivadas , Complemento C3/metabolismo , Proteínas Fetales/genética , Voluntarios Sanos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Riñón , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Primates , Proteínas Tirosina Quinasas/genética
4.
Gastroenterology ; 159(3): 1019-1035.e22, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32446697

RESUMEN

BACKGROUND & AIMS: Pancreatic tumor cells release small extracellular vesicles (sEVs, exosomes) that contain lipids and proteins, RNA, and DNA molecules that might promote formation of metastases. It is not clear what cargo these vesicles contain and how they are released. Protein kinase D1 (PRKD1) inhibits cell motility and is believed to be dysregulated in pancreatic ductal adenocarcinomas. We investigated whether it regulates production of sEVs in pancreatic cancer cells and their ability to form premetastatic niches for pancreatic cancer cells in mice. METHODS: We analyzed data from UALCAN and human pancreatic tissue microarrays to compare levels of PRKD1 between tumor and nontumor tissues. We studied mice with pancreas-specific disruption of Prkd1 (PRKD1KO mice), mice that express oncogenic KRAS (KC mice), and KC mice with disruption of Prkd1 (PRKD1KO-KC mice). Subcutaneous xenograft tumors were grown in NSG mice from Panc1 cells; some mice were then given injections of sEVs. Pancreata and lung tissues from mice were analyzed by histology, immunohistochemistry, and/or quantitative polymerase chain reaction; we performed nanoparticle tracking analysis of plasma sEVs. The Prkd1 gene was disrupted in Panc1 cells using CRISPR-Cas9 or knocked down with small hairpin RNAs, or PRKD1 activity was inhibited with the selective inhibitor CRT0066101. Pancreatic cancer cell lines were analyzed by gene-expression microarray, quantitative polymerase chain reaction, immunoblot, and immunofluorescence analyses. sEVs secreted by Panc1 cell lines were analyzed by flow cytometry, transmission electron microscopy, and mass spectrometry. RESULTS: Levels of PRKD1 were reduced in human pancreatic ductal adenocarcinoma tissues compared with nontumor tissues. PRKD1KO-KC mice developed more pancreatic intraepithelial neoplasia, at a faster rate, than KC mice, and had more lung metastases and significantly shorter average survival time. Serum from PRKD1KO-KC mice had increased levels of sEVs compared with KC mice. Pancreatic cancer cells with loss or inhibition of PRKD1 increased secretion of sEVs; loss of PRKD1 reduced phosphorylation of its substrate, cortactin, resulting in increased F-actin levels at the plasma membrane. sEVs from cells with loss or reduced expression of PRKD1 had altered content, and injection of these sEVs into mice increased metastasis of xenograft tumors to lung, compared with sEVs from pancreatic cells that expressed PRKD1. PRKD1-deficient pancreatic cancer cells showed increased loading of integrin α6ß4 into sEVs-a process that required CD82. CONCLUSIONS: Human pancreatic ductal adenocarcinoma has reduced levels of PRKD1 compared with nontumor pancreatic tissues. Loss of PRKD1 results in reduced phosphorylation of cortactin in pancreatic cancer cell lines, resulting in increased in F-actin at the plasma membrane and increased release of sEVs, with altered content. These sEVs promote metastasis of xenograft and pancreatic tumors to lung in mice.


Asunto(s)
Carcinoma Ductal Pancreático/secundario , Vesículas Extracelulares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pancreáticas/patología , Proteína Quinasa C/deficiencia , Animales , Carcinogénesis/patología , Carcinoma Ductal Pancreático/sangre , Línea Celular Tumoral , Movimiento Celular , Conjuntos de Datos como Asunto , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Pulmón/patología , Neoplasias Pulmonares/sangre , Ratones , Ratones Noqueados , Invasividad Neoplásica/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Páncreas/patología , Neoplasias Pancreáticas/sangre , Fosforilación , Cultivo Primario de Células , Proteína Quinasa C/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Clin Invest ; 128(11): 5056-5072, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30320600

RESUMEN

Dysregulated intestinal epithelial apoptosis initiates gut injury, alters the intestinal barrier, and can facilitate bacterial translocation leading to a systemic inflammatory response syndrome (SIRS) and/or multi-organ dysfunction syndrome (MODS). A variety of gastrointestinal disorders, including inflammatory bowel disease, have been linked to intestinal apoptosis. Similarly, intestinal hyperpermeability and gut failure occur in critically ill patients, putting the gut at the center of SIRS pathology. Regulation of apoptosis and immune-modulatory functions have been ascribed to Thirty-eight-negative kinase 1 (TNK1), whose activity is regulated merely by expression. We investigated the effect of TNK1 on intestinal integrity and its role in MODS. TNK1 expression induced crypt-specific apoptosis, leading to bacterial translocation, subsequent septic shock, and early death. Mechanistically, TNK1 expression in vivo resulted in STAT3 phosphorylation, nuclear translocation of p65, and release of IL-6 and TNF-α. A TNF-α neutralizing antibody partially blocked development of intestinal damage. Conversely, gut-specific deletion of TNK1 protected the intestinal mucosa from experimental colitis and prevented cytokine release in the gut. Finally, TNK1 was found to be deregulated in the gut in murine and porcine trauma models and human inflammatory bowel disease. Thus, TNK1 might be a target during MODS to prevent damage in several organs, notably the gut.


Asunto(s)
Proteínas Fetales/metabolismo , Enfermedades Inflamatorias del Intestino/enzimología , Intestinos/enzimología , Insuficiencia Multiorgánica/enzimología , Traumatismo Múltiple/enzimología , Proteínas Tirosina Quinasas/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/enzimología , Animales , Modelos Animales de Enfermedad , Femenino , Proteínas Fetales/genética , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Interleucina-6/genética , Interleucina-6/metabolismo , Intestinos/patología , Ratones , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/patología , Traumatismo Múltiple/complicaciones , Traumatismo Múltiple/genética , Traumatismo Múltiple/patología , Proteínas Tirosina Quinasas/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Porcinos , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/patología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Oncotarget ; 8(44): 77474-77488, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-29100402

RESUMEN

Lately, the HSP90 client serine/threonine kinase STK33 emerged to be required by cancer cells for their viability and proliferation. However, its mechanistic contribution to carcinogenesis is not clearly understood. Here we report that elevated STK33 expression correlates with advanced stages of human pancreatic and colorectal carcinomas. Impaired proliferation and augmented apoptosis associated with genetic abrogation of STK33 were paralleled by decreased vascularization in tumor xenografts. In line with this, ectopic STK33 not only promoted tumor growth after pharmacologic inhibition of HSP90 using structurally divergent small molecules currently in clinical development, but also restored blood vessel formation in vivo. Mechanistic studies demonstrated that HSP90-stabilized STK33 interacts with and regulates hypoxia-driven accumulation and activation of HIF-1α as well as secretion of VEGF-A in hypoxic cancer cells. In addition, our study reveals a putative cooperation between STK33 and other HSP90 client protein kinases involved in molecular and cellular events through which cancer cells ensure their survival by securing the oxygen and nutrient supply. Altogether, our findings indicate that STK33 interferes with signals from hypoxia and HSP90 to promote tumor angiogenesis and tumor growth.

7.
Mol Biol Cell ; 25(3): 324-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24336522

RESUMEN

Pancreatic cancer cell invasion, metastasis, and angiogenesis are major challenges for the development of novel therapeutic strategies. Protein kinase D (PKD) isoforms are involved in controlling tumor cell motility, angiogenesis, and metastasis. In particular PKD2 expression is up-regulated in pancreatic cancer, whereas PKD1 expression is lowered. We report that both kinases control pancreatic cancer cell invasive properties in an isoform-specific manner. PKD2 enhances invasion in three-dimensional extracellular matrix (3D-ECM) cultures by stimulating expression and secretion of matrix metalloproteinases 7 and 9 (MMP7/9), by which MMP7 is likely to act upstream of MMP9. Knockdown of MMP7/9 blocks PKD2-mediated invasion in 3D-ECM assays and in vivo using tumors growing on chorioallantois membranes. Furthermore, MMP9 enhances PKD2-mediated tumor angiogenesis by releasing extracellular matrix-bound vascular endothelial growth factor A, increasing its bioavailability and angiogenesis. Of interest, specific knockdown of PKD1 in PKD2-expressing pancreatic cancer cells further enhanced the invasive properties in 3D-ECM systems by generating a high-motility phenotype. Loss of PKD1 thus may be beneficial for tumor cells to enhance their matrix-invading abilities. In conclusion, we define for the first time PKD1 and 2 isoform-selective effects on pancreatic cancer cell invasion and angiogenesis, in vitro and in vivo, addressing PKD isoform specificity as a major factor for future therapeutic strategies.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/patología , Proteína Quinasa C/genética , Proteínas Quinasas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Membrana Corioalantoides/citología , Matriz Extracelular , Células HEK293 , Células HeLa , Humanos , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Metástasis de la Neoplasia/genética , Neovascularización Patológica/genética , Proteína Quinasa D2 , Interferencia de ARN , ARN Interferente Pequeño , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
J Cell Sci ; 125(Pt 9): 2148-59, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22344252

RESUMEN

Cell migration and invasion are largely dependent on the complex organization of the various cytoskeletal components. Whereas the role of actin filaments and microtubules in cell motility is well established, the role of intermediate filaments in this process is incompletely understood. Organization and structure of the keratin cytoskeleton, which consists of heteropolymers of at least one type 1 and one type 2 intermediate filament, are in part regulated by post-translational modifications. In particular, phosphorylation events influence the properties of the keratin network. Sphingosylphosphorylcholine (SPC) is a bioactive lipid with the exceptional ability to change the organization of the keratin cytoskeleton, leading to reorganization of keratin filaments, increased elasticity, and subsequently increased migration of epithelial tumor cells. Here we investigate the signaling pathways that mediate SPC-induced keratin reorganization and the role of keratin phosphorylation in this process. We establish that the MEK-ERK signaling cascade regulates both SPC-induced keratin phosphorylation and reorganization in human pancreatic and gastric cancer cells and identify Ser431 in keratin 8 as the crucial residue whose phosphorylation is required and sufficient to induce keratin reorganization and consequently enhanced migration of human epithelial tumor cells.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Filamentos Intermedios/metabolismo , Queratina-8/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Citoesqueleto/efectos de los fármacos , Citoesqueleto/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Filamentos Intermedios/efectos de los fármacos , Filamentos Intermedios/genética , Queratina-8/genética , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
9.
Mol Biol Cell ; 21(6): 1011-22, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20089835

RESUMEN

Protein kinase D (PKD) isoenzymes regulate the formation of transport carriers from the trans-Golgi network (TGN) that are en route to the plasma membrane. The PKD C1a domain is required for the localization of PKDs at the TGN. However, the precise mechanism of how PKDs are recruited to the TGN is still elusive. Here, we report that ADP-ribosylation factor (ARF1), a small GTPase of the Ras superfamily and a key regulator of secretory traffic, specifically interacts with PKD isoenzymes. ARF1, but not ARF6, binds directly to the second cysteine-rich domain (C1b) of PKD2, and precisely to Pro275 within this domain. Pro275 in PKD2 is not only crucial for the PKD2-ARF1 interaction but also for PKD2 recruitment to and PKD2 function at the TGN, namely, protein transport to the plasma membrane. Our data suggest a novel model in which ARF1 recruits PKD2 to the TGN by binding to Pro275 in its C1b domain followed by anchoring of PKD2 in the TGN membranes via binding of its C1a domain to diacylglycerol. Both processes are critical for PKD2-mediated protein transport.


Asunto(s)
Factor 1 de Ribosilacion-ADP/metabolismo , Isoenzimas/metabolismo , Prolina/metabolismo , Proteínas Quinasas/metabolismo , Transporte de Proteínas/fisiología , Red trans-Golgi/metabolismo , Factor 1 de Ribosilacion-ADP/genética , Línea Celular , Humanos , Isoenzimas/genética , Proteína Quinasa D2 , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
Cell Signal ; 21(2): 253-63, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19038333

RESUMEN

Protein Kinase D (PKD) has been implicated in the regulation of actin turnover at the leading edge, invasion and migration. In particular, a complex between cortactin, paxillin and PKD in the invadopodia of invasive breast cancer cells has been described earlier, but so far this complex remained ill defined. Here we have investigated the possible role of PKD as a cortactin kinase. Using a mass spectrometric approach, we found that PKD phosphorylates cortactin on Ser 298 in the 6th cortactin repeat region and on Ser 348, right before the helical-proline rich domain of cortactin. We developed phosphospecific antibodies against these phosphorylated sequences, and used them as tools to follow the in vivo phosphorylation of cortactin by PKD. Examination of cortactin phosphorylation kinetics revealed that Ser 298 serves as a priming site for subsequent phosphorylation of Ser 348. Src, a well-known cortactin kinase, strongly potentiated the in vivo PKD mediated cortactin phosphorylation. This Src effect is neither mediated by pre-phosphorylation of cortactin nor by activation of PKD by Src. Phosphorylation of cortactin by PKD does not affect its subcellular localization, nor does it affect its translocation to podosomes or membrane ruffles. Moreover, there was no effect of PKD mediated cortactin phosphorylation on EGF receptor degradation and LPA induced migration. Taken together, these data establish cortactin as a novel PKD substrate and reveal a novel connection between Src and PKD.


Asunto(s)
Cortactina/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Secuencia de Aminoácidos , Línea Celular , Cortactina/inmunología , Células HeLa , Humanos , Inmunoprecipitación , Espectrometría de Masas , Microscopía Fluorescente , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA