Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Oral Maxillofac Surg ; 82(9): 1038-1051.e1, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38825321

RESUMEN

BACKGROUND: The advantages of virtual surgical planning (VSP) for orthognathic surgery are clear. Previous studies have evaluated in-house VSP; however, few fully digital, in-house protocols for orthognathic surgery have been studied. PURPOSE: The purpose of this study was to evaluate the difference between the virtual surgical plan and actual surgical outcome for orthognathic surgery using a fully digital, in-house VSP workflow. STUDY DESIGN, SETTING, SAMPLE: This is a prospective cohort study from September 2020 to November 2022 of patients at the Victoria General Hospital in Halifax, NS, Canada who underwent bimaxillary orthognathic surgery. Patients were excluded if they had previously undergone orthognathic surgery or were diagnosed with a craniofacial syndrome. MAIN OUTCOME VARIABLES: The primary outcome variables were the mean 3-dimensional (3D) (Euclidean) distance error, as well as mean error and mean absolute error in the transverse (x axis), vertical (y axis), and anterior-posterior (z axis) dimensions. COVARIATES: Covariates included age, sex, and surgical sequence (mandible-first or maxilla-first). ANALYSES: The primary outcome was tested using Z and t critical value confidence intervals. The P value was set at .05. The 3D distance error for mandible-first and maxilla-first groups was compared using a 2-sample t-test as well as analysis of variance. RESULTS: The study sample included 52 subjects (24 males and 28 females) with a mean age of 27.7 (± 12.1) years. Forty three subjects underwent mandible-first surgery and 9 maxilla-first surgery. The mean absolute distance error was largest in the anterior-posterior dimension for all landmarks (except posterior nasal spine, left condyle, and gonion) and exceeded the threshold for clinical acceptability (2 mm) in 16 of 23 landmarks. Additionally, mean distance error in the anterior-posterior dimension was negative for all landmarks, indicating deficient movement in that direction. The effect of surgical sequence on 3D distance error was not statistically significant (P = .37). CONCLUSION AND RELEVANCE: In general, the largest contributor to mean 3D distance error was deficient movement in the anterior-posterior direction. Otherwise, mean absolute distance error in the vertical and transverse dimensions was clinically acceptable (< 2 mm). These findings were felt to be valuable for treatment planning purposes when using a fully digital, in-house VSP workflow.


Asunto(s)
Procedimientos Quirúrgicos Ortognáticos , Cirugía Asistida por Computador , Flujo de Trabajo , Humanos , Masculino , Femenino , Estudios Prospectivos , Procedimientos Quirúrgicos Ortognáticos/métodos , Adulto , Cirugía Asistida por Computador/métodos , Imagenología Tridimensional/métodos , Planificación de Atención al Paciente , Maxilar/cirugía , Maxilar/diagnóstico por imagen , Adulto Joven , Adolescente
2.
Sci Rep ; 10(1): 11650, 2020 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-32661249

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Cells ; 9(6)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32599788

RESUMEN

Ca2+ is an integral mediator of intracellular signaling, impacting almost every aspect of cellular life. The Ca2+-conducting transporters located on the endoplasmic reticulum (ER) membrane shoulder the responsibility of constructing the global Ca2+ signaling landscape. These transporters gate the ER Ca2+ release and uptake, sculpt signaling duration and intensity, and compose the Ca2+ signaling rhythm to accommodate a plethora of biological activities. In this review, we explore the mechanisms of activation and functional regulation of ER Ca2+ transporters in the establishment of Ca2+ homeostasis. We also contextualize the aberrant alterations of these transporters in carcinogenesis, presenting Ca2+-based therapeutic interventions as a means to tackle malignancies.


Asunto(s)
Señalización del Calcio/genética , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Humanos
4.
Adv Exp Med Biol ; 1131: 747-770, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31646533

RESUMEN

The pioneering work of Richard Altman on the presence of mitochondria in cells set in motion a field of research dedicated to uncovering the secrets of the mitochondria. Despite limitations in studying the structure and function of the mitochondria, advances in our understanding of this organelle prompted the development of potential treatments for various diseases, from neurodegenerative conditions to muscular dystrophy and cancer. As the powerhouses of the cell, the mitochondria represent the essence of cellular life and as such, a selective advantage for cancer cells. Much of the function of the mitochondria relies on Ca2+ homeostasis and the presence of effective Ca2+ signaling to maintain the balance between mitochondrial function and dysfunction and subsequently, cell survival. Ca2+ regulates the mitochondrial respiration rate which in turn increases ATP synthesis, but too much Ca2+ can also trigger the mitochondrial apoptosis pathway; however, cancer cells have evolved mechanisms to modulate mitochondrial Ca2+ influx and efflux in order to sustain their metabolic demand and ensure their survival. Therefore, targeting the mitochondrial Ca2+ signaling involved in the bioenergetic and apoptotic pathways could serve as potential approaches to treat cancer patients. This chapter will review the role of Ca2+ signaling in mediating the function of the mitochondria and its involvement in health and disease with special focus on the pathophysiology of cancer.


Asunto(s)
Señalización del Calcio , Calcio , Mitocondrias , Neoplasias , Apoptosis , Calcio/metabolismo , Señalización del Calcio/fisiología , Homeostasis , Humanos , Mitocondrias/fisiología , Neoplasias/fisiopatología
5.
J Proteome Res ; 19(2): 708-718, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31884793

RESUMEN

The efficacy of oncolytic viruses (OVs), such as reovirus, is dictated by host immune responses, including those mediated by the pro- versus anti-inflammatory macrophages. As such, a detailed understanding of the interaction between reovirus and different macrophage types is critical for therapeutic efficacy. To explore reovirus-macrophage interactions, we performed tandem mass tag (TMT)-based quantitative temporal proteomics on mouse bone marrow-derived macrophages (BMMs) generated with two cytokines, macrophage colony stimulating factor (M-CSF) and granulocytic-macrophage colony stimulating factor (GM-CSF), representing anti- and proinflammatory macrophages, respectively. We quantified 6863 proteins across five time points in duplicate, comparing M-CSF (M-BMM) and GM-CSF (GM-BMM) in response to OV. We find that GM-BMMs have lower expression of key intrinsic proteins that facilitate an antiviral immune response, express higher levels of reovirus receptor protein JAM-A, and are more susceptible to oncolytic reovirus infection compared to M-BMMs. Interestingly, although M-BMMs are less susceptible to reovirus infection and subsequent cell death, they initiate an antireovirus adaptive T cell immune response comparable to that of GM-BMMs. Taken together, these data describe distinct proteome differences between these two macrophage populations in terms of their ability to mount antiviral immune responses.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Factor Estimulante de Colonias de Macrófagos , Animales , Médula Ósea , Células de la Médula Ósea , Células Cultivadas , Ratones , Proteoma
6.
Cell Calcium ; 82: 102053, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31279156

RESUMEN

Gastric cancer is a multifactorial disease associated with a combination of and environmental factors. Each year, one million new gastric cancer cases are diagnosed worldwide and two-thirds end up losing the battle with this devastating disease. Currently, surgery represents the only effective treatment option for patients with early stage tumors. However, the asymptomatic phenotype of this disease during the early stages poses as a significant limiting factor to diagnosis and often renders treatments ineffective. To address these issues, scientists are focusing on personalized medicine and discovering new ways to treat cancer patients. Emerging therapeutic options include the transient receptor potential (TRP) channels. Since their discovery, TRP channels have been shown to contribute significantly to the pathophysiology of various cancers, including gastric cancer. This review will summarize the current knowledge about gastric cancer and provide a synopsis of recent advancements on the role and involvement of TRP channels in gastric cancer as well as a discussion of the benefits of targeting TPR channel in the clinical management of gastric cancer.


Asunto(s)
Neoplasias Gástricas/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Medicina de Precisión , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patología
7.
Cell Physiol Biochem ; 52(4): 742-757, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30933439

RESUMEN

BACKGROUND/AIMS: The oxidative stress sensor transient receptor potential melastatin-2 (TRPM2) ion channel has recently gained attention in many types of cancer. The lung tissue is highly susceptible to oxidative stress-mediated injury and diseases; therefore, we aimed to determine whether TRPM2 plays an essential role in protecting lung cancer cells from oxidative damage while promoting cancer cell survival and metastasis. METHODS: We used two non-small cell lung (NSCLC) cell lines A549 and H1299 as a lung cancer model. We investigated the functional expression of TRPM2 using electrophysiology, qRT-PCR and Western blots. CFSE and flow cytometry were used to study TRPM2 role in proliferation, cell cycle and apoptosis. Gap closure chambers and Three-Tiered Chemotaxis Chamber were used to study the role of TRPM2 in metastasis. SCID mice were used to study the role of TRPM2 in lung tumor growth and metastasis. RESULTS: we demonstrate that TRPM2 is functionally expressed in NSCLC cells and that its downregulation significantly inhibits cell proliferation and promotes apoptosis. These results were concomitant with an induction in DNA damage and G2/M cell cycle arrest. TRPM2 silencing inhibits also lung cancer cells invasion ability and alters EMT processes. Mechanistically, TRPM2 downregulation causes an increase in the intracellular levels of reactive oxygen (ROS) and nitrogen (RNS) species, which in turn causes DNA damage and JNK activation leading to G2/M arrest, and an ultimate cell death. Finally, TRPM2 downregulation suppresses the growth of human lung tumour xenograft in SCID mice and TRPM2 depleted tumours exhibited a significant reduction in the mRNA expression level of EMT markers compared to the control tumors. CONCLUSION: Our data provide new insights on the functional expression of TRPM2 in lung cancer, its essential role in tumour growth and metastasis through the control of JNK signaling pathway, and that TRPM2 could be exploited for targeted lung cancer therapies.


Asunto(s)
Apoptosis , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPM/metabolismo , Animales , Antracenos/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Puntos de Control de la Fase M del Ciclo Celular , Sistema de Señalización de MAP Quinasas , Ratones , Ratones SCID , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética
8.
Cell Calcium ; 79: 80-88, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30889511

RESUMEN

The triple-negative breast cancer (TNBC) that comprises approximately 10%-20% of breast cancers is an aggressive subtype lacking effective therapeutics. Among various signaling pathways, mTORC1 and purinergic signals have emerged as potentially fruitful targets for clinical therapy of TNBC. Unfortunately, drugs targeting these signaling pathways do not successfully inhibit the progression of TNBC, partially due to the fact that these signaling pathways are essential for the function of all types of cells. In this study, we report that TRPML1 is specifically upregulated in TNBCs and that its genetic downregulation and pharmacological inhibition suppress the growth of TNBC. Mechanistically, we demonstrate that TRPML1 regulates TNBC development, at least partially, through controlling mTORC1 activity and the release of lysosomal ATP. Because TRPML1 is specifically activated by cellular stresses found in tumor microenvironments, antagonists of TRPML1 could represent anticancer drugs with enhanced specificity and potency. Our findings are expected to have a major impact on drug targeting of TNBCs.


Asunto(s)
Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal , Canales de Potencial de Receptor Transitorio/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Calcio/metabolismo , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Canales de Potencial de Receptor Transitorio/deficiencia , Neoplasias de la Mama Triple Negativas/patología
9.
Sci Rep ; 9(1): 4182, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862883

RESUMEN

Transient Receptor Potential Melastatin-2 (TRPM2) ion channel is emerging as a great therapeutic target in many types of cancer, including gastric cancer - a major health threat of cancer related-death worldwide. Our previous study demonstrated the critical role of TRPM2 in gastric cancer cells bioenergetics and survival; however, its role in gastric cancer metastasis, the major cause of patient death, remains unknown. Here, using molecular and functional assays, we demonstrate that TRPM2 downregulation significantly inhibits the migration and invasion abilities of gastric cancer cells, with a significant reversion in the expression level of metastatic markers. These effects were concomitant with decreased Akt and increased PTEN activities. Finally, TRPM2 silencing resulted in deregulation of metastatic markers and abolished the tumor growth ability of AGS gastric cancer cells in NOD/SCID mice. Taken together, our results provide compelling evidence on the important function of TRPM2 in the modulation of gastric cancer cell invasion likely through controlling the PTEN/Akt pathway.


Asunto(s)
Movimiento Celular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Canales Catiónicos TRPM/metabolismo , Animales , Calcio/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Citosol/metabolismo , Regulación hacia Abajo , Activación Enzimática , Transición Epitelial-Mesenquimal , Silenciador del Gen , Células HEK293 , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica
10.
Sci Rep ; 8(1): 14091, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30237490

RESUMEN

Cancer dissemination is initiated by the movement of cells into the vasculature which has been reported to be triggered by EMT (epithelial to mesenchymal transition). Cellular dissemination also requires proteases that remodel the extracellular matrix. The protease, plasmin is a prominent player in matrix remodeling and invasion. Despite the contribution of both EMT and the plasminogen activation (PA) system to cell dissemination, these processes have never been functionally linked. We reveal that canonical Smad-dependent TGFß1 signaling and FOXC2-mediated PI3K signaling in cells undergoing EMT reciprocally modulate plasminogen activation partly by regulating the plasminogen receptor, S100A10 and the plasminogen activation inhibitor, PAI-1. Plasminogen activation and plasminogen-dependent invasion were more prominent in epithelial-like cells and were partly dictated by the expression of S100A10 and PAI-1.


Asunto(s)
Anexina A2/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Plasminógeno/metabolismo , Proteínas S100/metabolismo , Línea Celular , Fibrinolisina/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo
11.
Mol Oncol ; 12(11): 1895-1916, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30009399

RESUMEN

Pancreatic cancer is arguably the deadliest cancer type. The efficacy of current therapies is often hindered by the inability to predict patient outcome. As such, the development of tools for early detection and risk prediction is key for improving outcome and quality of life. Here, we introduce the plasminogen receptor S100A10 as a novel predictive biomarker and a driver of pancreatic tumor growth and invasion. We demonstrated that S100A10 mRNA and protein are overexpressed in human pancreatic tumors compared to normal ducts and nonductal stroma. S100A10 mRNA and methylation status were predictive of overall survival and recurrence-free survival across multiple patient cohorts. S100A10 expression was driven by promoter methylation and the oncogene KRAS. S100A10 knockdown reduced surface plasminogen activation, invasiveness, and in vivo growth of pancreatic cancer cell lines. These findings delineate the clinical and functional contribution of S100A10 as a biomarker in pancreatic cancer.


Asunto(s)
Anexina A2/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas , Proteínas S100/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Tasa de Supervivencia
12.
Cell Calcium ; 72: 91-103, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29748137

RESUMEN

Lysosomes serve as the control centre for cellular clearance. These membrane-bound organelles receive biomolecules destined for degradation from intracellular and extracellular pathways; thus, facilitating the production of energy and shaping the fate of the cell. At the base of their functionality are the lysosomal ion channels which mediate the function of the lysosome through the modulation of ion influx and efflux. Ion channels form pores in the membrane of lysosomes and allow the passage of ions, a seemingly simple task which harbours the potential of overthrowing the cell's stability. Considered the master regulators of ion homeostasis, these integral membrane proteins enable the proper operation of the lysosome. Defects in the structure or function of these ion channels lead to the development of lysosomal storage diseases, neurodegenerative diseases and cancer. Although more than 50 years have passed since their discovery, lysosomes are not yet fully understood, with their ion channels being even less well characterized. However, significant improvements have been made in the development of drugs targeted against these ion channels as a means of combating diseases. In this review, we will examine how Ca2+, K+, Na+ and Cl- ion channels affect the function of the lysosome, their involvement in hereditary and spontaneous diseases, and current ion channel-based therapies.


Asunto(s)
Canales Iónicos/metabolismo , Lisosomas/metabolismo , Animales , Enfermedad , Humanos , Canales Iónicos/química , Modelos Biológicos
13.
J Biol Chem ; 293(10): 3637-3650, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29343514

RESUMEN

A lack of effective treatment is one of the main factors contributing to gastric cancer-related death. Discovering effective targets and understanding their underlying anti-cancer mechanism are key to achieving the best response to treatment and to limiting side effects. Although recent studies have shown that the cation channel transient receptor potential melastatin-2 (TRPM2) is crucial for cancer cell survival, the exact mechanism remains unclear, limiting its therapeutic potential. Here, using molecular and functional assays, we investigated the role of TRPM2 in survival of gastric cancer cells. Our results indicated that TRPM2 knockdown in AGS and MKN-45 cells decreases cell proliferation and enhances apoptosis. We also observed that the TRPM2 knockdown impairs mitochondrial metabolism, indicated by a decrease in basal and maximal mitochondrial oxygen consumption rates and ATP production. These mitochondrial defects coincided with a decrease in autophagy and mitophagy, indicated by reduced levels of autophagy- and mitophagy-associated proteins (i.e. ATGs, LC3A/B II, and BNIP3). Moreover, we found that TRPM2 modulates autophagy through a c-Jun N-terminal kinase (JNK)-dependent and mechanistic target of rapamycin-independent pathway. We conclude that in the absence of TRPM2, down-regulation of the JNK-signaling pathway impairs autophagy, ultimately causing the accumulation of damaged mitochondria and death of gastric cancer cells. Of note, by inhibiting cell proliferation and promoting apoptosis, the TRPM2 down-regulation enhanced the efficacy of paclitaxel and doxorubicin in gastric cancer cells. Collectively, we provide compelling evidence that TRPM2 inhibition may benefit therapeutic approaches for managing gastric cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Apoptosis , Autofagia , Mitofagia , Proteínas de Neoplasias/metabolismo , Neoplasias Gástricas/metabolismo , Canales Catiónicos TRPM/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Registros Electrónicos de Salud , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Fosforilación Oxidativa/efectos de los fármacos , Paclitaxel/farmacología , Interferencia de ARN , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Análisis de Supervivencia , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética
14.
J Proteome Res ; 16(9): 3391-3406, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28768414

RESUMEN

Myeloid cells play a central role in the context of viral eradication, yet precisely how these cells differentiate throughout the course of acute infections is poorly understood. In this study, we have developed a novel quantitative temporal in vivo proteomics (QTiPs) platform to capture proteomic signatures of temporally transitioning virus-driven myeloid cells directly in situ, thus taking into consideration host-virus interactions throughout the course of an infection. QTiPs, in combination with phenotypic, functional, and metabolic analyses, elucidated a pivotal role for inflammatory CD11b+, Ly6G-, Ly6Chigh-low cells in antiviral immune response and viral clearance. Most importantly, the time-resolved QTiPs data set showed the transition of CD11b+, Ly6G-, Ly6Chigh-low cells into M2-like macrophages, which displayed increased antigen-presentation capacities and bioenergetic demands late in infection. We elucidated the pivotal role of myeloid cells in virus clearance and show how these cells phenotypically, functionally, and metabolically undergo a timely transition from inflammatory to M2-like macrophages in vivo. With respect to the growing appreciation for in vivo examination of viral-host interactions and for the role of myeloid cells, this study elucidates the use of quantitative proteomics to reveal the role and response of distinct immune cell populations throughout the course of virus infection.


Asunto(s)
Interacciones Huésped-Patógeno , Macrófagos/metabolismo , Células Mieloides/metabolismo , Proteómica/métodos , Infecciones por Reoviridae/genética , Animales , Antígenos Ly/genética , Antígenos Ly/inmunología , Biomarcadores/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Diferenciación Celular , Proliferación Celular , Eliminación de Gen , Regulación de la Expresión Génica , Ontología de Genes , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Células Mieloides/inmunología , Células Mieloides/virología , Orthoreovirus de los Mamíferos/crecimiento & desarrollo , Orthoreovirus de los Mamíferos/patogenicidad , Receptores CCR2/genética , Receptores CCR2/inmunología , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/virología , Transducción de Señal , Factores de Tiempo
15.
Viruses ; 7(12): 6506-25, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26690204

RESUMEN

Dendritic cells (DCs) are specialized antigen-presenting cells that have a notable role in the initiation and regulation of innate and adaptive immune responses. In the context of cancer, appropriately activated DCs can induce anti-tumor immunity by activating innate immune cells and tumor-specific lymphocytes that target cancer cells. However, the tumor microenvironment (TME) imposes different mechanisms that facilitate the impairment of DC functions, such as inefficient antigen presentation or polarization into immunosuppressive DCs. These tumor-associated DCs thus fail to initiate tumor-specific immunity, and indirectly support tumor progression. Hence, there is increasing interest in identifying interventions that can overturn DC impairment within the TME. Many reports thus far have studied oncolytic viruses (OVs), viruses that preferentially target and kill cancer cells, for their capacity to enhance DC-mediated anti-tumor effects. Herein, we describe the general characteristics of DCs, focusing on their role in innate and adaptive immunity in the context of the TME. We also examine how DC-OV interaction affects DC recruitment, OV delivery, and anti-tumor immunity activation. Understanding these roles of DCs in the TME and OV infection is critical in devising strategies to further harness the anti-tumor effects of both DCs and OVs, ultimately enhancing the efficacy of OV-based oncotherapy.


Asunto(s)
Células Dendríticas/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Inmunidad Celular
16.
J Immunol ; 194(9): 4397-412, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825443

RESUMEN

Tumor-associated immunosuppression aids cancer cells to escape immune-mediated attack and subsequent elimination. Recently, however, many oncolytic viruses, including reovirus, have been reported to overturn such immunosuppression and promote the development of a clinically desired antitumor immunity, which is known to promote favorable patient outcomes. Contrary to this existing paradigm, in this article we demonstrate that reovirus augments tumor-associated immunosuppression immediately following its therapeutic administration. Our data show that reovirus induces preferential differentiation of highly suppressive CD11b(+), Gr-1(+), Ly6C(high) myeloid cells from bone marrow hematopoietic progenitor cells. Furthermore, reovirus administration in tumor-bearing hosts drives time-dependent recruitment of CD11b(+), Gr-1(+), Ly6C(high) myeloid cells in the tumor milieu, which is further supported by virus-induced increased expression of numerous immune factors involved in myeloid-derived suppressor cell survival and trafficking. Most importantly, CD11b(+), Gr-1(+), Ly6C(high) myeloid cells specifically potentiate the suppression of T cell proliferation and are associated with the absence of IFN-γ response in the tumor microenvironment early during oncotherapy. Considering that the qualitative traits of a specific antitumor immunity are largely dictated by the immunological events that precede its development, our findings are of critical importance and must be considered while devising complementary interventions aimed at promoting the optimum efficacy of oncolytic virus-based anticancer immunotherapies.


Asunto(s)
Vectores Genéticos , Inmunomodulación , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/inmunología , Virus Oncolíticos , Fenotipo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular , Quimiotaxis/inmunología , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/inmunología , Ratones , Células Mieloides/citología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Receptores de Quimiocina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA