Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
FASEB J ; 37(6): e22995, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37219526

RESUMEN

Immuno-oncology (IO)-based therapies such as checkpoint inhibitors, bi-specific antibodies, and CAR-T-cell therapies have shown significant success in the treatment of several cancer indications. However, these therapies can result in the development of severe adverse events, including cytokine release syndrome (CRS). Currently, there is a paucity of in vivo models that can evaluate dose-response relationships for both tumor control and CRS-related safety issues. We tested an in vivo PBMC humanized mouse model to assess both treatment efficacy against specific tumors and the concurrent cytokine release profiles for individual human donors after treatment with a CD19xCD3 bispecific T-cell engager (BiTE). Using this model, we evaluated tumor burden, T-cell activation, and cytokine release in response to bispecific T-cell-engaging antibody in humanized mice generated with different PBMC donors. The results show that PBMC engrafted NOD-scid Il2rgnull mice lacking expression of mouse MHC class I and II (NSG-MHC-DKO mice) and implanted with a tumor xenograft predict both efficacy for tumor control by CD19xCD3 BiTE and stimulated cytokine release. Moreover, our findings indicate that this PBMC-engrafted model captures variability among donors for tumor control and cytokine release following treatment. Tumor control and cytokine release were reproducible for the same PBMC donor in separate experiments. The PBMC humanized mouse model described here is a sensitive and reproducible platform that identifies specific patient/cancer/therapy combinations for treatment efficacy and development of complications.


Asunto(s)
Leucocitos Mononucleares , Linfocitos T , Humanos , Animales , Ratones , Ratones Endogámicos NOD , Resultado del Tratamiento , Síndrome de Liberación de Citoquinas , Citocinas , Modelos Animales de Enfermedad , Ratones Noqueados , Ratones SCID
2.
Toxicol Pathol ; 51(4): 205-215, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37578155

RESUMEN

The evaluation of changes in the immune system serves to determine the efficacy and potential immunotoxicologic effects of new products under development. Toxicologic pathologists play critical roles in identifying immune system changes that drive the immunosafety determination. Standard pathology evaluations of therapies and chemicals remain similar; however, biopharmaceutical therapies have moved from simply affecting the immune system to being specifically developed to modify the immune system, which can impact interpretation. Recent explosive growth in immunomodulatory therapies presents a challenge to the toxicologic pathologist, toxicologist, and regulatory reviewer in terms of evaluating the clinical relevance and potential adversity of immune system changes. Beyond the recognition of such changes, there is an increasing expectation to evaluate, describe, and interpret how therapies affect complex immune system pathways for both immunomodulatory therapies and non-immunomodulatory drugs with off-target immunotoxic effects. In this opinion piece, considerations regarding immune system evaluation, the current landscape of immunomodulatory therapies, a brief description of immunotoxicologic (and immunopathologic) endpoints, the importance of integrating such immunosafety data, and relevance to adversity determination are discussed. Importantly, we describe how the current paradigm of determining adversity for immune system changes may be challenging or insufficient and propose a harmonized and flexible approach for assessing adversity.

3.
Toxicol Pathol ; 47(2): 121-128, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30651043

RESUMEN

GlaxoSmithKline has recently made significant organizational changes to its nonclinical safety, drug metabolism and pharmacokinetic, and laboratory animal science/veterinary functions, with the goal to increase our focus on scientific partnership with the discovery part of the organization. One specific change was bringing together pathologists and comparative medicine veterinarians and scientists into a single functional unit. We describe our early activities (assessing our capabilities and gaps, external benchmarking, listening to our discovery partners, redesigning some of our working practices) aimed at implementing these changes. In addition, early on we held a Discovery Engagement Workshop attended by all pathologists and comparative medicine veterinarians and scientists, as well as selected discovery scientists. The purpose of this workshop was to share learnings from the above activities and devise plans aimed at achieving our overall goal of functional integration: driving pathobiology expertise into drug discovery and increasing the human (translational) relevance of experimental data. This review describes the new organizational structure, the workshop activities, and implementation plans; updates our progress; and considers the opportunity for a pan-industry network of discovery-focused pathologists and comparative medicine veterinarians and scientists.


Asunto(s)
Descubrimiento de Drogas/métodos , Industria Farmacéutica/organización & administración , Personal de Laboratorio , Patólogos , Animales , Evaluación Preclínica de Medicamentos/métodos , Humanos , Patología , Veterinarios
4.
Cell Tissue Res ; 370(2): 275-283, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28766045

RESUMEN

We previously found that transgenic mice overexpressing MMTV-FLAG-hPAD2 (PAD2OE) developed spontaneous skin lesions, with a subset of these lesions progressing to invasive squamous cell carcinoma (SCC). The goal of this report was to better understand the potential mechanisms by which PAD2 overexpression promotes skin cancer. Here, PAD2OE mice were treated with the carcinogen, 9,10-dimethyl-1,2-benzanthracene and with O-tetradecanoylphorbol-13-acetate and then scored for papilloma formation. Additionally, tumor sections were evaluated for evidence of tumor cell invasion and inflammation. We found that the total number of papillomas was significantly increased in PAD2OE mice compared to controls. Histopathologic analysis of the lesions found that in PAD2OE skin tumors progressed to invasive SCC more frequently than controls. Additionally, we found that PAD2OE lesions were highly inflamed, with a dense inflammatory cell infiltrate and an associated increase in nuclear phospho-STAT3 (signal transducer and activator of transcription 3) in the transgenic tumors. These data suggest that overexpression of the hPAD2 transgene in the epidermis increases the malignant conversion rate of benign tumors by promoting an inflammatory microenvironment.


Asunto(s)
Inflamación/genética , Papiloma/genética , Desiminasas de la Arginina Proteica/genética , Neoplasias Cutáneas/genética , Regulación hacia Arriba , 9,10-Dimetil-1,2-benzantraceno , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Carcinogénesis/patología , Carcinógenos , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/complicaciones , Inflamación/patología , Masculino , Ratones , Ratones Transgénicos , Papiloma/inducido químicamente , Papiloma/complicaciones , Papiloma/patología , Arginina Deiminasa Proteína-Tipo 2 , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/complicaciones , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol
5.
Proc Natl Acad Sci U S A ; 110(29): 11851-6, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818587

RESUMEN

Peptidylarginine deiminase 4 (PAD4) is a Ca(2+)-dependent enzyme that converts arginine and methylarginine residues to citrulline, with histone proteins being among its best-described substrates to date. However, the biological function of this posttranslational modification, either in histones or in nonhistone proteins, is poorly understood. Here, we show that PAD4 recognizes, binds, and citrullinates glycogen synthase kinase-3ß (GSK3ß), both in vitro and in vivo. Among other functions, GSK3ß is a key regulator of transcription factors involved in tumor progression, and its dysregulation has been associated with progression of human cancers. We demonstrate that silencing of PAD4 in breast cancer cells leads to a striking reduction of nuclear GSK3ß protein levels, increased TGF-ß signaling, induction of epithelial-to-mesenchymal transition, and production of more invasive tumors in xenograft assays. Moreover, in breast cancer patients, reduction of PAD4 and nuclear GSK3ß is associated with increased tumor invasiveness. We propose that PAD4-mediated citrullination of GSK3ß is a unique posttranslational modification that regulates its nuclear localization and thereby plays a critical role in maintaining an epithelial phenotype. We demonstrate a dynamic and previously unappreciated interplay between histone-modifying enzymes, citrullination of nonhistone proteins, and epithelial-to-mesenchymal transition.


Asunto(s)
Citrulina/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Hidrolasas/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Ionóforos de Calcio , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Células MCF-7 , Espectrometría de Masas , Microscopía de Interferencia , Mutagénesis Sitio-Dirigida , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
6.
Proc Natl Acad Sci U S A ; 109(25): 9786-91, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22665775

RESUMEN

Multipotent adipose-derived stem cells (ASCs) are increasingly used for regenerative purposes such as soft tissue reconstruction following mastectomy; however, the ability of tumors to commandeer ASC functions to advance tumor progression is not well understood. Through the integration of physical sciences and oncology approaches we investigated the capability of tumor-derived chemical and mechanical cues to enhance ASC-mediated contributions to tumor stroma formation. Our results indicate that soluble factors from breast cancer cells inhibit adipogenic differentiation while increasing proliferation, proangiogenic factor secretion, and myofibroblastic differentiation of ASCs. This altered ASC phenotype led to varied extracellular matrix (ECM) deposition and contraction thereby enhancing tissue stiffness, a characteristic feature of breast tumors. Increased stiffness, in turn, facilitated changes in ASC behavior similar to those observed with tumor-derived chemical cues. Orthotopic mouse studies further confirmed the pathological relevance of ASCs in tumor progression and stiffness in vivo. In summary, altered ASC behavior can promote tumorigenesis and, thus, their implementation for regenerative therapy should be carefully considered in patients previously treated for cancer.


Asunto(s)
Tejido Adiposo/citología , Neoplasias de la Mama/terapia , Trasplante de Células Madre , Animales , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Matriz Extracelular , Femenino , Humanos , Ratones , Trasplante de Neoplasias
7.
Int J Urol ; 21(3): 319-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24033563

RESUMEN

OBJECTIVES: To study the relationship between myosin light chain phosphorylation of the detrusor muscle and spontaneous smooth muscle contractions in a rabbit model of partial outlet obstruction. METHODS: New Zealand white rabbit urinary bladders were partially obstructed for 2 weeks. Rabbits were euthanized, detrusor muscle strips were hung on a force transducer and spontaneous activity was measured at varying concentrations (0-0.03 µM/L) of the Rho-kinase inhibitors GSK 576371 or 0.01 µM/L Y27632. Basal myosin light chain phosphorylation was measured by 2-D gel electrophoresis in control and GSK 576371-treated strips. RESULTS: Both drugs suppressed the force of spontaneous contractions, whereas GSK 576371 had a more profound effect on the frequency of the contractions. The IC50 values for the inhibition of frequency and force of spontaneous contractions were 0.17 µM/L and 0.023 µM/L for GSK 576371, respectively. The compound significantly decreased the basal myosin light chain phosphorylation from 28.0 ± 3.9% to 13.5 ± 1.9% (P < 0.05). At 0.01 µM/L, GSK 576371 inhibited spontaneous bladder overactivity by 50%, but inhibited carbachol-elicited contractions force by just 25%. CONCLUSIONS: These data suggest that Rho-kinase regulation of myosin light chain phosphorylation contributes to the spontaneous detrusor activity induced by obstruction. This finding could have therapeutic implications by providing another therapeutic option for myogenic, overactive bladder.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Cadenas Ligeras de Miosina/metabolismo , Vejiga Urinaria Hiperactiva/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Masculino , Datos de Secuencia Molecular , Fosforilación/efectos de los fármacos , Conejos , Obstrucción del Cuello de la Vejiga Urinaria/complicaciones , Vejiga Urinaria Hiperactiva/etiología
8.
J Allergy Clin Immunol ; 132(4): 811-20.e1-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23768572

RESUMEN

BACKGROUND: Asthma is a predominantly TH2 cell-dominated inflammatory disease characterized by airway inflammation and a major public health concern affecting millions of persons. The Tec family tyrosine kinase IL-2-inducible T-cell kinase (Itk) is primarily expressed in T cells and critical for the function and differentiation of TH cells. Itk(-/-) mice have a defective TH2 response and are not susceptible to allergic asthma. OBJECTIVE: We sought to better understand the role of Itk signaling in TH differentiation programs and in the development and molecular pathology of allergic asthma. METHODS: Using a murine model of allergic airway inflammation, we dissected the role of Itk in regulating TH cell differentiation through genetic ablation of critical genes, chromatin immunoprecipitation assays, and house dust mite-driven allergic airway inflammation. RESULTS: Peripheral naive Itk(-/-) CD4(+) T cells have substantially increased transcripts and expression of the prototypic TH1 genes Eomesodermin, IFN-γ, T-box transcription factor (T-bet), and IL-12Rß1. Removal of IFN-γ on the Itk(-/-) background rescues expression of TH2-related genes in TH cells and allergic airway inflammation in Itk(-/-) mice. Furthermore, small hairpin RNA-mediated knockdown of Itk in human peripheral blood T cells results in increased expression of mRNA for IFN-γ and T-bet and reduction in expression of IL-4. CONCLUSION: Our results indicate that Itk signals suppress the expression of IFN-γ in naive CD4(+) T cells, which in a positive feed-forward loop regulates the expression of TH1 factors, such as T-bet and Eomesodermin, and suppress development of TH2 cells and allergic airway inflammation.


Asunto(s)
Asma/inmunología , Hipersensibilidad/inmunología , Inflamación/inmunología , Interferón gamma/efectos de los fármacos , Proteínas Tirosina Quinasas/inmunología , Células Th2/inmunología , Animales , Asma/etiología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Inmunoprecipitación de Cromatina , Femenino , Humanos , Hipersensibilidad/etiología , Inflamación/etiología , Interferón gamma/metabolismo , Masculino , Ratones , Proteínas Tirosina Quinasas/metabolismo , Pyroglyphidae/inmunología , Células TH1/citología , Células TH1/inmunología , Células Th2/citología
9.
Mol Imaging Biol ; 25(3): 528-540, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36266600

RESUMEN

PURPOSE: The presence and functional competence of intratumoral CD8+ T cells is often a barometer for successful immunotherapeutic responses in cancer. Despite this understanding and the extensive number of clinical-stage immunotherapies focused on potentiation (co-stimulation) or rescue (checkpoint blockade) of CD8+ T cell antitumor activity, dynamic biomarker strategies are often lacking. To help fill this gap, immuno-PET nuclear imaging has emerged as a powerful tool for in vivo molecular imaging of antibody targeting. Here, we took advantage of immuno-PET imaging using 89Zr-IAB42M1-14, anti-mouse CD8 minibody, to characterize CD8+ T-cell tumor infiltration dynamics following ICOS (inducible T-cell co-stimulator) agonist antibody treatment alone and in combination with PD-1 blocking antibody in a model of mammary carcinoma. PROCEDURES: Female BALB/c mice with established EMT6 tumors received 10 µg, IP of either IgG control antibodies, ICOS agonist monotherapy, or ICOS/PD-1 combination therapy on days 0, 3, 5, 7, 9, 10, or 14. Imaging was performed at 24 and 48 h post IV dose of 89Zr IAB42M1-14. In addition to 89Zr-IAB42M1-14 uptake in tumor and tumor-draining lymph node (TDLN), 3D radiomic features were extracted from PET/CT images to identify treatment effects. Imaging mass cytometry (IMC) and immunohistochemistry (IHC) was performed at end of study. RESULTS: 89Zr-IAB42M1-14 uptake in the tumor was observed by day 11 and was preceded by an increase in the TDLN as early as day 4. The spatial distribution of 89Zr-IAB42M1-14 was more uniform in the drug treated vs. control tumors, which had spatially distinct tracer uptake in the periphery relative to the core of the tumor. IMC analysis showed an increased percentage of cytotoxic T cells in the ICOS monotherapy and ICOS/PD-1 combination group compared to IgG controls. Additionally, temporal radiomics analysis demonstrated early predictiveness of imaging features. CONCLUSION: To our knowledge, this is the first detailed description of the use of a novel immune-PET imaging technique to assess the kinetics of CD8+ T-cell infiltration into tumor and lymphoid tissues following ICOS agonist and PD-1 blocking antibody therapy. By demonstrating the capacity for increased spatial and temporal resolution of CD8+ T-cell infiltration across tumors and lymphoid tissues, these observations underscore the widespread potential clinical utility of non-invasive PET imaging for T-cell-based immunotherapy in cancer.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Ratones , Femenino , Linfocitos T CD8-positivos/patología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptor de Muerte Celular Programada 1 , Neoplasias/patología , Tomografía de Emisión de Positrones/métodos , Inmunoglobulina G , Línea Celular Tumoral , Proteína Coestimuladora de Linfocitos T Inducibles
10.
BMC Cancer ; 12: 500, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23110523

RESUMEN

BACKGROUND: We have recently reported that the expression of peptidylarginine deiminase 2 (PADI2) is regulated by EGF in mammary cancer cells and appears to play a role in the proliferation of normal mammary epithelium; however, the role of PADI2 in the pathogenesis of human breast cancer has yet to be investigated. Thus, the goals of this study were to examine whether PADI2 plays a role in mammary tumor progression, and whether the inhibition of PADI activity has anti-tumor effects. METHODS: RNA-seq data from a collection of 57 breast cancer cell lines was queried for PADI2 levels, and correlations with known subtype and HER2/ERBB2 status were evaluated. To examine PADI2 expression levels during breast cancer progression, the cell lines from the MCF10AT model were used. The efficacy of the PADI inhibitor, Cl-amidine, was tested in vitro using MCF10DCIS cells grown in 2D-monolayers and 3D-spheroids, and in vivo using MCF10DCIS tumor xenografts. Treated MCF10DCIS cells were examined by flow-cytometry to determine the extent of apoptosis and by RT2 Profiler PCR Cell Cycle Array to detect alterations in cell cycle associated genes. RESULTS: We show by RNA-seq that PADI2 mRNA expression is highly correlated with HER2/ERBB2 (p = 2.2 × 106) in luminal breast cancer cell lines. Using the MCF10AT model of breast cancer progression, we then demonstrate that PADI2 expression increases during the transition of normal mammary epithelium to fully malignant breast carcinomas, with a strong peak of PADI2 expression and activity being observed in the MCF10DCIS cell line, which models human comedo-DCIS lesions. Next, we show that a PADI inhibitor, Cl-amidine, strongly suppresses the growth of MCF10DCIS monolayers and tumor spheroids in culture. We then carried out preclinical studies in nude (nu/nu) mice and found that Cl-amidine also suppressed the growth of xenografted MCF10DCIS tumors by more than 3-fold. Lastly, we performed cell cycle array analysis of Cl-amidine treated and control MCF10DCIS cells, and found that the PADI inhibitor strongly affects the expression of several cell cycle genes implicated in tumor progression, including p21, GADD45α, and Ki67. CONCLUSION: Together, these results suggest that PADI2 may function as an important new biomarker for HER2/ERBB2+ tumors and that Cl-amidine represents a new candidate for breast cancer therapy.


Asunto(s)
Biomarcadores de Tumor/fisiología , Neoplasias de la Mama/enzimología , Hidrolasas/fisiología , Proteínas de Neoplasias/fisiología , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Perfilación de la Expresión Génica , Humanos , Hidrolasas/antagonistas & inhibidores , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica
11.
Lab Invest ; 89(7): 823-32, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19381130

RESUMEN

Normal urinary bladder function requires contraction and relaxation of the detrusor smooth muscle (DSM). The DSM undergoes compensatory hypertrophy in response to partial bladder outlet obstruction (PBOO) in both men and animal models. Following bladder hypertrophy, the bladder either retains its normal function (compensated) or becomes dysfunctional (decompensated) with increased voiding frequency and decreased void volume. We analyzed the contractile characteristics of DSM in a rabbit model of PBOO. The protein kinase C (PKC) agonist phorbol 12, 13-dibutyrate (PDBu) elicited similar levels of contraction of DSM strips from normal and compensated bladders. However, PDBu-induced contraction decreased significantly in DSM strips from decompensated bladders. The expression and activity of PKC-alpha were also lowest in decompensated bladders. The PKC-specific inhibitor bisindolylmaleimide-1 (Bis) blocked PDBu-induced contraction and PKC activity in all three groups. Moreover, the phosphorylation of the phosphoprotein inhibitor CPI-17 (a 17-kDa PKC-potentiated inhibitory protein of protein phosphatase-1) was diminished in DSM from the decompensated bladder, which would result in less inhibitory potency of CPI-17 on myosin light chain phosphatase activity and contribute to less contractility. Immunostaining revealed the colocalization of PKC and phosphorylated CPI-17 in the DSM and confirmed the decreases of these signaling proteins in the decompensated bladder. Our results show a differential PKC-mediated DSM contraction with corresponding alterations of PKC expression, activity and the phosphorylation of CPI-17. Our finding suggests a significant correlation between bladder function and PKC pathway. An impaired PKC pathway appears to be correlated with severe bladder dysfunction observed in decompensated bladders.


Asunto(s)
Músculo Liso/fisiopatología , Proteína Quinasa C/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/fisiopatología , Vejiga Urinaria/patología , Vejiga Urinaria/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Hipertrofia , Técnicas In Vitro , Indoles/farmacología , Masculino , Maleimidas/farmacología , Contracción Muscular , Proteínas Musculares/metabolismo , Músculo Liso/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación , Cloruro de Potasio/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Conejos , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología
12.
J Urol ; 182(5): 2497-503, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19765744

RESUMEN

PURPOSE: Partial bladder outlet obstruction in male rabbits causes detrusor smooth muscle hypertrophy and voiding dysfunction similar to that observed in men with benign prostate hyperplasia. Using this model, we analyzed the protein expression and ultrastructure of caveolae and the intermediate size filament in detrusor smooth muscle following partial bladder outlet obstruction induced hypertrophy. MATERIALS AND METHODS: Detrusor smooth muscle sections from bladder body were processed for immunofluorescence and electron microscopy. Western analysis was performed to determine the expression of caveolin isoform-1, 2 and 3, and intermediate size filament proteins. RESULTS: Detrusor smooth muscle cells from both normal and hypertrophied bladders contain orderly arrays of thick and thin myofilaments, interspersed with dense bodies. In addition, there was an increase in intermediate size filaments in the hypertrophic detrusor smooth muscle cells. The dense plaques in the inner membrane of hypertrophied detrusor smooth muscle were longer than those of the control. Detrusor smooth muscle from hypertrophied bladder revealed a decreased number of caveolae and a lack of their orderly distribution at the plasma membrane. Western blotting showed decreased expression of caveolin-1, 2 and 3 in hypertrophied detrusor smooth muscle. CONCLUSIONS: Caveolae serve as platforms for proteins and receptors that have a role in signal transduction. The decreased number of caveolae and caveolin protein expression in hypertrophied detrusor smooth muscle might contribute to alterations in signal transduction pathways that regulate the downstream effects of agonist induced contraction, including calcium sensitization, observed in obstructed bladder. In addition, the increased number of intermediate size filaments in the hypertrophied detrusor smooth muscle is likely to alter the cytoskeletal structure and affect the cellular transmission of passive and/or active force.


Asunto(s)
Caveolinas/biosíntesis , Músculo Liso/metabolismo , Músculo Liso/patología , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Animales , Caveolas , Hipertrofia , Técnicas In Vitro , Masculino , Microscopía Electrónica , Músculo Liso/ultraestructura , Conejos , Vejiga Urinaria/ultraestructura
13.
Biomaterials ; 224: 119489, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31546097

RESUMEN

While ductal carcinoma in situ (DCIS) is known as a precursor lesion to most invasive breast carcinomas, the mechanisms underlying this transition remain enigmatic. DCIS is typically diagnosed by the mammographic detection of microcalcifications (MC). MCs consisting of non-stoichiometric hydroxyapatite (HA) mineral are frequently associated with malignant disease, yet it is unclear whether HA can actively promote malignancy. To investigate this outstanding question, we compared phenotypic outcomes of breast cancer cells cultured in control or HA-containing poly(lactide-co-glycolide) (PLG) scaffolds. Exposure to HA mineral in scaffolds increased the expression of pro-tumorigenic interleukin-8 (IL-8) among transformed but not benign cells. Notably, MCF10DCIS.com cells cultured in HA scaffolds adopted morphological changes associated with increased invasiveness and exhibited increased motility that were dependent on IL-8 signaling. Moreover, MCF10DCIS.com xenografts in HA scaffolds displayed evidence of enhanced malignant progression relative to xenografts in control scaffolds. These experimental findings were supported by a pathological analysis of clinical DCIS specimens, which correlated the presence of MCs with increased IL-8 staining and ductal proliferation. Collectively, our work suggests that HA mineral may stimulate malignancy in preinvasive DCIS cells and validate PLG scaffolds as useful tools to study cell-mineral interactions.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Durapatita/farmacología , Minerales/farmacología , Modelos Biológicos , Ingeniería de Tejidos , Animales , Neoplasias de la Mama/complicaciones , Calcinosis/complicaciones , Carcinoma Intraductal no Infiltrante/complicaciones , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Interleucina-8/metabolismo , Ratones Desnudos , Invasividad Neoplásica , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Andamios del Tejido/química
14.
J Food Prot ; 69(9): 2263-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16995535

RESUMEN

In the manufacture of beef jerky, a thermal lethality step is followed by drying to prevent growth of pathogenic bacterial postprocessing contaminants on the finished product. Recent guidelines from the U.S. Department of Agriculture have raised the question of the maximum water activity (a(w)) in jerky products that will inhibit growth of pathogenic bacteria. The survival of the potential postprocessing contaminants Staphylococcus aureus and Listeria monocytogenes was evaluated on 15 vacuum-packaged beef jerky and related products with a(w) values ranging from 0.47 to 0.87, just below the 0.88 limit reported for anaerobic growth of S. aureus. Small individual product pieces were inoculated on the outer surface with five strains each of S. aureus and L. monocytogenes, repackaged under vacuum, and stored at room temperature (21 degrees C) for 4 weeks. Pathogen numbers were determined before storage and after 1 and 4 weeks. None of the 15 jerky products supported growth of either pathogen. Counts of S. aureus fell by 0.2 to 1.8 log CFU after 1 week of storage and by 0.6 to 5.3 log CFU after 4 weeks of storage. Numbers of L. monocytogenes fell by 0.6 to 4.7 log CFU and by 2.3 to 5.6 log CFU after 1 and 4 weeks of storage, respectively. Although factors other than a(w) may have some effect on pathogen survival, the results of the present study clearly support drying beef jerky to an a(w) of < or = 0.87 to ensure that bacterial pathogens cannot grow on vacuum-packaged product stored at room temperature.


Asunto(s)
Embalaje de Alimentos/métodos , Conservación de Alimentos/métodos , Listeria monocytogenes/crecimiento & desarrollo , Productos de la Carne/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Animales , Bovinos , Recuento de Colonia Microbiana , Humanos , Temperatura , Factores de Tiempo , Vacio , Agua/metabolismo
15.
Sci Transl Med ; 7(301): 301ra130, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26290412

RESUMEN

Obesity and extracellular matrix (ECM) density are considered independent risk and prognostic factors for breast cancer. Whether they are functionally linked is uncertain. We investigated the hypothesis that obesity enhances local myofibroblast content in mammary adipose tissue and that these stromal changes increase malignant potential by enhancing interstitial ECM stiffness. Indeed, mammary fat of both diet- and genetically induced mouse models of obesity were enriched for myofibroblasts and stiffness-promoting ECM components. These differences were related to varied adipose stromal cell (ASC) characteristics because ASCs isolated from obese mice contained more myofibroblasts and deposited denser and stiffer ECMs relative to ASCs from lean control mice. Accordingly, decellularized matrices from obese ASCs stimulated mechanosignaling and thereby the malignant potential of breast cancer cells. Finally, the clinical relevance and translational potential of our findings were supported by analysis of patient specimens and the observation that caloric restriction in a mouse model reduces myofibroblast content in mammary fat. Collectively, these findings suggest that obesity-induced interstitial fibrosis promotes breast tumorigenesis by altering mammary ECM mechanics with important potential implications for anticancer therapies.


Asunto(s)
Tejido Adiposo/metabolismo , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Matriz Extracelular/metabolismo , Obesidad/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Transformación Celular Neoplásica , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Obesos , Obesidad/complicaciones
16.
Cancer Res ; 74(21): 6306-17, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25213324

RESUMEN

Peptidylarginine deiminase 2 (PAD2/PADI2) has been implicated in various inflammatory diseases and, more recently, cancer. The goal of this study was to test the hypothesis that PAD2 promotes oncogenesis using a transgenic mouse model. We found that about 37% of transgenic mice overexpressing human FLAG-PAD2 downstream of the MMTV-LTR promoter develop spontaneous neoplastic skin lesions. Molecular and histopathologic analyses of the resulting lesions find that they contain increased levels of markers for invasion, inflammation, and epithelial-to-mesenchymal transition (EMT) and that a subset of the lesions progress to invasive squamous cell carcinoma (SCC). We then stably overexpressed FLAG-PAD2 in the human SCC cell line, A431, and found that the PAD2-overexpressing cells were more tumorigenic in vitro and also contained elevated levels of markers for inflammation and EMT. Collectively, these studies provide the first genetic evidence that PAD2 functions as an oncogene and suggest that PAD2 may promote tumor progression by enhancing inflammation within the tumor microenvironment.


Asunto(s)
Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Hidrolasas/biosíntesis , Neoplasias Cutáneas/genética , Animales , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal/genética , Humanos , Hidrolasas/genética , Ratones , Ratones Transgénicos , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica , Neoplasias Cutáneas/patología , Microambiente Tumoral
17.
Front Immunol ; 4: 67, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23508122

RESUMEN

PAD4-mediated hypercitrullination of histone H4 arginine 3 (H4R3) has been previously found to promote the formation of Neutrophil Extracellular Traps in inflamed tissues and the resulting histone H4 citrulline 3 (H4Cit3) modification is thought to play a key role in extracellular trap (ET) formation by promoting chromatin decondensation. In addition to neutrophils, macrophages have also recently been found to generate functional extracellular traps (METs). However, a role for PADs in ET formation in macrophages has not been previously described. Transcripts for PAD2 and PAD4 are found in mature macrophages and these cells can be induced to citrullinate proteins, thus raising the possibility that PADs may play a direct role in ET formation in macrophages via histone hypercitrullination. In breast and visceral white adipose tissue from obese patients, infiltrating macrophages are often seen to surround dead adipocytes forming characteristic "crown-like structures" (CLS) and the presence of these lesions is associated with increased levels of inflammatory mediators. In light of these observations, we have initiated studies to test whether PADs are expressed in CLS macrophages and whether these macrophages might form METs. Our preliminary findings show that PAD2 (and to a lesser extent, PAD4) is expressed in both in the macrophage cell line (RAW 264.7) and in CLS lesions. Additionally, we provide evidence that macrophage-derived extracellular histones are seen around presumptive macrophages within CLS lesions and that these histones contain the H4Cit3 modification. These initial findings support our hypothesis that obesity-induced adipose tissue inflammation promotes the formation of METs within CLS lesions via PAD-mediated histone hypercitrullination. Subsequent studies are underway to further validate these findings and to investigate the role in PAD-mediated MET formation in CLS function in the mammary gland.

18.
Cancer Res ; 73(23): 7079-89, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24121485

RESUMEN

Glioblastoma multiforme contains a subpopulation of cancer stem-like cells (CSC) believed to underlie tumorigenesis and therapeutic resistance. Recent studies have localized CSCs in this disease adjacent to endothelial cells (EC) in what has been termed a perivascular niche, spurring investigation into the role of EC-CSC interactions in glioblastoma multiforme pathobiology. However, these studies have been limited by a lack of in vitro models of three-dimensional disease that can recapitulate the relevant conditions of the niche. In this study, we engineered a scaffold-based culture system enabling brain endothelial cells to form vascular networks. Using this system, we showed that vascular assembly induces CSC maintenance and growth in vitro and accelerates tumor growth in vivo through paracrine interleukin (IL)-8 signaling. Relative to conventional monolayers, endothelial cells cultured in this three-dimensional system not only secreted enhanced levels of IL-8 but also induced CSCs to upregulate the IL-8 cognate receptors CXCR1 and CXCR2, which collectively enhanced CSC migration, growth, and stemness properties. CXCR2 silencing in CSCs abolished the tumor-promoting effects of endothelial cells in vivo, confirming a critical role for this signaling pathway in GMB pathogenesis. Together, our results reveal synergistic interactions between endothelial cells and CSCs that promote the malignant properties of CSCs in an IL-8-dependent manner. Furthermore, our findings underscore the relevance of tissue-engineered cell culture platforms to fully analyze signaling mechanisms in the tumor microenvironment.


Asunto(s)
Glioblastoma/patología , Interleucina-8/fisiología , Células Madre Neoplásicas/metabolismo , Nicho de Células Madre/fisiología , Animales , Vasos Sanguíneos/patología , Técnicas de Cultivo de Célula , Células Cultivadas , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Glioblastoma/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones SCID , Comunicación Paracrina/fisiología , Andamios del Tejido , Microambiente Tumoral/fisiología
19.
J Bone Miner Res ; 28(11): 2357-67, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23649605

RESUMEN

Bone metastasis, the leading cause of breast cancer-related deaths, is characterized by bone degradation due to increased osteoclastic activity. In contrast, mechanical stimulation in healthy individuals upregulates osteoblastic activity, leading to new bone formation. However, the effect of mechanical loading on the development and progression of metastatic breast cancer in bone remains unclear. Here, we developed a new in vivo model to investigate the role of skeletal mechanical stimuli on the development and osteolytic capability of secondary breast tumors. Specifically, we applied compressive loading to the tibia following intratibial injection of metastatic breast cancer cells (MDA-MB231) into the proximal compartment of female immunocompromised (SCID) mice. In the absence of loading, tibiae developed histologically-detectable tumors with associated osteolysis and excessive degradation of the proximal bone tissue. In contrast, mechanical loading dramatically reduced osteolysis and tumor formation and increased tibial cancellous mass due to trabecular thickening. These loading effects were similar to the baseline response we observed in non-injected SCID mice. In vitro mechanical loading of MDA-MB231 in a pathologically relevant 3D culture model suggested that the observed effects were not due to loading-induced tumor cell death, but rather mediated via decreased expression of genes interfering with bone homeostasis. Collectively, our results suggest that mechanical loading inhibits the growth and osteolytic capability of secondary breast tumors after their homing to the bone, which may inform future treatment of breast cancer patients with advanced disease.


Asunto(s)
Neoplasias Óseas/fisiopatología , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Osteólisis/patología , Osteólisis/fisiopatología , Tibia/fisiopatología , Animales , Neoplasias Óseas/complicaciones , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/secundario , Resorción Ósea/complicaciones , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/diagnóstico por imagen , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones SCID , Osteólisis/complicaciones , Osteólisis/diagnóstico por imagen , Tibia/patología , Andamios del Tejido/química , Soporte de Peso , Microtomografía por Rayos X
20.
Biochem Res Int ; 2012: 895343, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23019525

RESUMEN

The peptidylarginine deiminases (PADs) are a family of posttranslational modification enzymes that catalyze the conversion of positively charged protein-bound arginine and methylarginine residues to the uncharged, nonstandard amino acid citrulline. This enzymatic activity is referred to as citrullination or, alternatively, deimination. Citrullination can significantly affect biochemical pathways by altering the structure and function of target proteins. Five mammalian PAD family members (PADs 1-4 and 6) have been described and show tissue-specific distribution. Recent reviews on PADs have focused on their role in autoimmune diseases. Here, we will discuss the potential role of PADs in tumor progression and tumor-associated inflammation. In the context of cancer, increasing clinical evidence suggests that PAD4 (and possibly PAD2) has important roles in tumor progression. The link between PADs and cancer is strengthened by recent findings showing that treatment of cell lines and mice with PAD inhibitors significantly suppresses tumor growth and, interestingly, inflammatory symptoms. At the molecular level, transcription factors, coregulators, and histones are functional targets for citrullination by PADs, and citrullination of these targets can affect gene expression in multiple tumor cell lines. Next generation isozyme-specific PAD inhibitors may have therapeutic potential to regulate both the inflammatory tumor microenvironment and tumor cell growth.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA