Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 70(3): 633-656, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32865605

RESUMEN

Adoptive transfer of Bispecific antibody Armed activated T cells (BATs) showed promising anti-tumor activity in clinical trials in solid tumors. The cytotoxic activity of BATs occurs upon engagement with tumor cells via the bispecific antibody (BiAb) bridge, which stimulates BATs to release cytotoxic molecules, cytokines, chemokines, and other signaling molecules extracellularly. We hypothesized that the release of BATs Induced Tumor-Targeting Effectors (TITE) by this complex interaction of T cells, bispecific antibody, and tumor cells may serve as a potent anti-tumor and immune-activating immunotherapeutic approach. In a 3D tumorsphere model, TITE showed potent cytotoxic activity against multiple breast cancer cell lines compared to control conditioned media (CM): Tumor-CM (T-CM), BATs-CM (B-CM), BiAb Armed PBMC-CM (BAP-CM) or PBMC-CM (P-CM). Multiplex cytokine analysis showed high levels of Th1 cytokines and chemokines; phospho-protein signaling array data suggest that the prominent JAK1/STAT1 pathway may be responsible for the induction and release of Th1 cytokines/chemokines in TITE. In xenograft breast cancer models, IV injections of 10× concentrated TITE (3×/week for 3 weeks; 150 µl TITE/injection) was able to inhibit tumor growth significantly (ICR/scid, p < 0.003; NSG p < 0.008) compared to the control mice. We tested the key components of the TITE for immune activating and anti-tumor activity individually and in combinations, the combination of IFN-γ, TNF-α and MIP-1ß recapitulates the key activities of the TITE. In summary, master mix of active components of BATs-Tumor complex-derived TITE can provide a clinically controllable cell-free platform to target various tumor types regardless of the heterogeneous nature of the tumor cells and mutational tumor.


Asunto(s)
Citotoxicidad Inmunológica , Inmunomodulación , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores , Línea Celular Tumoral , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Ratones , Neoplasias/diagnóstico , Neoplasias/terapia , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Genes Dev ; 27(19): 2086-98, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24065739

RESUMEN

Serine and cysteine cathepsin (Cts) proteases are an important class of intracellular and pericellular enzymes mediating multiple aspects of tumor development. Emblematic of these is CtsB, reported to play functionally significant roles during pancreatic islet and mammary carcinogenesis. CtsC, on the other hand, while up-regulated during pancreatic islet carcinogenesis, lacks functional significance in mediating neoplastic progression in that organ. Given that protein expression and enzymatic activity of both CtsB and CtsC are increased in numerous tumors, we sought to understand how tissue specificity might factor into their functional significance. Thus, whereas others have reported that CtsB regulates metastasis of mammary carcinomas, we found that development of squamous carcinomas occurs independently of CtsB. In contrast to these findings, our studies found no significant role for CtsC during mammary carcinogenesis but revealed squamous carcinogenesis to be functionally dependent on CtsC. In this context, dermal/stromal fibroblasts and bone marrow-derived cells expressed increased levels of enzymatically active CtsC that regulated the complexity of infiltrating immune cells in neoplastic skin, development of angiogenic vasculature, and overt squamous cell carcinoma growth. These studies highlight the important contribution of tissue/microenvironment context to solid tumor development and indicate that tissue specificity defines functional significance for these two members of the cysteine protease family.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/fisiopatología , Catepsina C/metabolismo , Neoplasias Cutáneas/fisiopatología , Animales , Catepsina B/genética , Catepsina B/metabolismo , Catepsina C/genética , Línea Celular Tumoral , Quimasas/metabolismo , Femenino , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Leucocitos/metabolismo , Neoplasias Mamarias Animales/fisiopatología , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética , Elastasa Pancreática/metabolismo
3.
Cancer Metastasis Rev ; 38(1-2): 103-112, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31069574

RESUMEN

The glycolytic phenotype of the Warburg effect is associated with acidification of the tumor microenvironment. In this review, we describe how acidification of the tumor microenvironment may increase the invasive and degradative phenotype of cancer cells. As a template of an extracellular acidic microenvironment that is linked to proteolysis, we use the resorptive pit formed between osteoclasts and bone. We describe similar changes that have been observed in cancer cells in response to an acidic microenvironment and that are associated with proteolysis and invasive and metastatic phenotypes. This includes consideration of changes observed in the intracellular trafficking of vesicles, i.e., lysosomes and exosomes, and in specialized regions of the membrane, i.e., invadopodia and caveolae. Cancer-associated cells are known to affect what is generally referred to as tumor proteolysis but little direct evidence for this being regulated by acidosis; we describe potential links that should be verified.


Asunto(s)
Acidosis/metabolismo , Acidosis/patología , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral/fisiología , Animales , Humanos , Proteolisis
4.
Cancer Metastasis Rev ; 38(3): 445-454, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31605250

RESUMEN

Cells grown in three dimensions (3D) within natural extracellular matrices or synthetic scaffolds more closely recapitulate the phenotype of those cells within tissues in regard to normal developmental and pathobiological processes. This includes degradation of the surrounding stroma as the cells migrate and invade through the matrices. As 3D cultures of tumor cells predict efficacy of, and resistance to, a wide variety of cancer therapies, we employed tissue-engineering approaches to establish 3D pathomimetic avatars of human breast cancer cells alone and in the context of both their cellular and pathochemical microenvironments. We have shown that we can localize and quantify key parameters of malignant progression by live-cell imaging of the 3D avatars over time (4D). One surrogate for changes in malignant progression is matrix degradation, which can be localized and quantified by our live-cell proteolysis assay. This assay is predictive of changes in spatio-temporal and dynamic interactions among the co-cultured cells and changes in viability, proliferation, and malignant phenotype. Furthermore, our live-cell proteolysis assay measures the effect of small-molecule inhibitors of proteases and kinases, neutralizing or blocking antibodies to cytokines and photodynamic therapy on malignant progression. We suggest that 3D/4D pathomimetic avatars in combination with our live-cell proteolysis assays will be a useful preclinical screening platform for cancer therapies. Our ultimate goal is to develop 3D/4D avatars from an individual patient's cancer in which we can screen "personalized medicine" therapies using changes in proteolytic activity to quantify therapeutic efficacy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Animales , Neoplasias de la Mama/diagnóstico por imagen , Carcinoma Ductal de Mama/diagnóstico por imagen , Carcinoma Intraductal no Infiltrante/diagnóstico por imagen , Técnicas de Cultivo de Célula/métodos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Microscopía Confocal , Imagen Óptica/métodos , Proteolisis , Microambiente Tumoral
5.
J Mammary Gland Biol Neoplasia ; 24(1): 1-15, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30056557

RESUMEN

About one fourth of all newly identified cases of breast carcinoma are diagnoses of breast ductal carcinoma in situ (DCIS). Since we cannot yet distinguish DCIS cases that would remain indolent from those that may progress to life-threatening invasive ductal carcinoma (IDC), almost all women undergo aggressive treatment. In order to allow for more rational individualized treatment, we and others are developing in vitro models to identify and validate druggable pathways that mediate the transition of DCIS to IDC. These models range from conventional two-dimensional (2D) monolayer cultures on plastic to 3D cultures in natural or synthetic matrices. Some models consist solely of DCIS cells, either cell lines or primary cells. Others are co-cultures that include additional cell types present in the normal or cancerous human breast. The 3D co-culture models more accurately mimic structural and functional changes in breast architecture that accompany the transition of DCIS to IDC. Mechanistic studies of the dynamic and temporal changes associated with this transition are facilitated by adapting the in vitro models to engineered microfluidic platforms. Ultimately, the goal is to create in vitro models that can serve as a reproducible preclinical screen for testing therapeutic strategies that will reduce progression of DCIS to IDC. This review will discuss the in vitro models that are currently available, as well as the progress that has been made using them to understand DCIS pathobiology.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Cultivo Primario de Células/métodos , Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Línea Celular Tumoral , Técnicas de Cocultivo/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control
6.
Adv Exp Med Biol ; 1152: 401-411, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31456196

RESUMEN

Understanding breast cancer cell proteolysis and migration is crucial for developing novel therapies to prevent local and distant metastases. Human cancer cells utilize many biological functions comparable to those observed during embryogenesis conferring the cancer cells with survival advantages. One such advantage is the ability to secrete proteases into the tumor microenvironment in order to remodel the extracellular matrix to facilitate migration. These proteases degrade the extracellular matrix, which initially functions as a barrier to cancer cell escape from their site of origin. The extracellular matrix also functions as a reservoir for growth factors that can be released by the secreted proteases and thereby further aid tumor growth and progression. Other survival advantages of tumor cells include: the ability to utilize multiple modes of motility, thrive in acidic microenvironments, and the tumor cell's ability to hijack stromal and immune cells to foster their own migration and survival. In order to reduce metastasis, we must focus our efforts on addressing the survival advantages that tumor cells have acquired.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Proteolisis , Matriz Extracelular , Femenino , Humanos , Péptido Hidrolasas/metabolismo , Microambiente Tumoral
7.
J Pathol ; 243(4): 496-509, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29086922

RESUMEN

Mammography screening has increased the detection of early pre-invasive breast cancers, termed ductal carcinoma in situ (DCIS), increasing the urgency of identifying molecular regulators of invasion as prognostic markers to predict local relapse. Using the MMTV-PyMT breast cancer model and pharmacological protease inhibitors, we reveal that cysteine cathepsins have important roles in early-stage tumorigenesis. To characterize the cell-specific roles of cathepsins in early invasion, we developed a DCIS-like model, incorporating an immortalized myoepithelial cell line (N1ME) that restrained tumor cell invasion in 3D culture. Using this model, we identified an important myoepithelial-specific function of the cysteine cathepsin inhibitor stefin A in suppressing invasion, whereby targeted stefin A loss in N1ME cells blocked myoepithelial-induced suppression of breast cancer cell invasion. Enhanced invasion observed in 3D cultures with N1ME stefin A-low cells was reliant on cathepsin B activation, as addition of the small molecule inhibitor CA-074 rescued the DCIS-like non-invasive phenotype. Importantly, we confirmed that stefin A was indeed abundant in myoepithelial cells in breast tissue. Use of a 138-patient cohort confirmed that myoepithelial stefin A (cystatin A) is abundant in normal breast ducts and low-grade DCIS but reduced in high-grade DCIS, supporting myoepithelial stefin A as a candidate marker of lower risk of invasive relapse. We have therefore identified myoepithelial cell stefin A as a suppressor of early tumor invasion and a candidate marker to distinguish patients who are at low risk of developing invasive breast cancer, and can therefore be spared further treatment. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Movimiento Celular , Cistatina A/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Catepsina B/antagonistas & inhibidores , Catepsina B/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Cistatina A/genética , Inhibidores de Cisteína Proteinasa/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Humanos , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/patología , Ratones , Invasividad Neoplásica , Interferencia de ARN , Transducción de Señal , Transfección , Microambiente Tumoral , Proteínas Supresoras de Tumor/genética
8.
Breast Cancer Res ; 19(1): 56, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506312

RESUMEN

BACKGROUND: The breast tumor microenvironment regulates progression of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC). However, it is unclear how interactions between breast epithelial and stromal cells can drive this progression and whether there are reliable microenvironmental biomarkers to predict transition of DCIS to IDC. METHODS: We used xenograft mouse models and a 3D pathomimetic model termed mammary architecture and microenvironment engineering (MAME) to study the interplay between human breast myoepithelial cells (MEPs) and cancer-associated fibroblasts (CAFs) on DCIS progression. RESULTS: Our results show that MEPs suppress tumor formation by DCIS cells in vivo even in the presence of CAFs. In the in vitro MAME model, MEPs reduce the size of 3D DCIS structures and their degradation of extracellular matrix. We further show that the tumor-suppressive effects of MEPs on DCIS are linked to inhibition of urokinase plasminogen activator (uPA)/urokinase plasminogen activator receptor (uPAR)-mediated proteolysis by plasminogen activator inhibitor 1 (PAI-1) and that they can lessen the tumor-promoting effects of CAFs by attenuating interleukin 6 (IL-6) signaling pathways. CONCLUSIONS: Our studies using MAME are, to our knowledge, the first to demonstrate a divergent interplay between MEPs and CAFs within the DCIS tumor microenvironment. We show that the tumor-suppressive actions of MEPs are mediated by PAI-1, uPA and its receptor, uPAR, and are sustained even in the presence of the CAFs, which themselves enhance DCIS tumorigenesis via IL-6 signaling. Identifying tumor microenvironmental regulators of DCIS progression will be critical for defining a robust and predictive molecular signature for clinical use.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Interleucina-6/genética , Inhibidor 1 de Activador Plasminogénico/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Animales , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal de Mama/patología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteoma/genética , Análisis de Matrices Tisulares , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Sens Actuators B Chem ; 247: 92-97, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28970651

RESUMEN

In the effort of developing micro-electrochemical sensors, the miniaturization of reference electrodes has been a challenging task. In this paper, a flexible micro reference electrode with an internal electrolyte reservoir is reported. This new device is based on a unique microfabricated parylene tube structure, which is filled with Cl- rich electrolyte, into which a 50 µm diameter silver (Ag) wire covered with a 7.4 µm thick silver chloride (AgCl) layer is inserted. The distal end of the tube is filled with potassium chloride (KCl) saturated agarose gel. The Ag wire, thick AgCl layer, and internal electrolyte reservoir lead to a long operation time and a stable reference voltage. The drift over a 10-hour period has been found to be less than 2 mV. The total operation time of the device has exceeded 100 hours. Furthermore, the compatibility with microfabrication allows the integration of other components, leading to truly miniaturized electrochemical sensors or sensing systems. To prove this, we demonstrated a pH sensor by combining the reference electrode and an iridium oxide electrode monolithically integrated on the surface of the parylene tube.

10.
Breast Cancer Res Treat ; 154(2): 251-62, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26502410

RESUMEN

Photodynamic therapy (PDT) is a minimally invasive, FDA-approved therapy for treatment of endobronchial and esophageal cancers that are accessible to light. Inflammatory breast cancer (IBC) is an aggressive and highly metastatic form of breast cancer that spreads to dermal lymphatics, a site that would be accessible to light. IBC patients have a relatively poor survival rate due to lack of targeted therapies. The use of PDT is underexplored for breast cancers but has been proposed for treatment of subtypes for which a targeted therapy is unavailable. We optimized and used a 3D mammary architecture and microenvironment engineering (MAME) model of IBC to examine the effects of PDT using two treatment protocols. The first protocol used benzoporphyrin derivative monoacid A (BPD) activated at doses ranging from 45 to 540 mJ/cm(2). The second PDT protocol used two photosensitizers: mono-L-aspartyl chlorin e6 (NPe6) and BPD that were sequentially activated. Photokilling by PDT was assessed by live-dead assays. Using a MAME model of IBC, we have shown a significant dose-response in photokilling by BPD-PDT. Sequential activation of NPe6 followed by BPD is more effective in photokilling of tumor cells than BPD alone. Sequential activation at light doses of 45 mJ/cm(2) for each agent resulted in >90 % cell death, a response only achieved by BPD-PDT at a dose of 360 mJ/cm(2). Our data also show that effects of PDT on a volumetric measurement of 3D MAME structures reflect efficacy of PDT treatment. Our study is the first to demonstrate the potential of PDT for treating IBC.


Asunto(s)
Neoplasias Inflamatorias de la Mama/patología , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Caspasa 3 , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Técnicas In Vitro , Neoplasias Inflamatorias de la Mama/terapia , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Técnicas de Cultivo de Tejidos , Microambiente Tumoral
11.
Biol Chem ; 396(3): 277-81, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25205719

RESUMEN

Cathepsin B has been demonstrated to be involved in several proteolytic processes that support tumor progression and metastasis and neurodegeneration. To further clarify its role, defined monoclonal antibodies are needed. As the primary structure of human cathepsin B is almost identical to that of the mouse, cathepsin B-deficient mice were used in a novel approach for generating such antibodies, providing the chance of an increased immune response to the antigen, human cathepsin B. Thirty clones were found to produce cathepsin B-specific antibodies. Seven of these antibodies were used to detect cathepsin B in MCF10-DCIS human breast cancer cells by immunocytochemistry and immunoblotting. Five different binding sites were identified by epitope mapping giving the opportunity to combine these antibodies in oligoclonal antibody mixtures for an improved detection of cathepsin B.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Catepsina B/deficiencia , Catepsina B/inmunología , Secuencia de Aminoácidos , Animales , Catepsina B/química , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Ratones , Datos de Secuencia Molecular
12.
BMC Cancer ; 15: 584, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26268945

RESUMEN

BACKGROUND: Ductal carcinoma in situ (DCIS) is a non-obligate precursor lesion of invasive breast cancer in which approximately half the patients will progress to invasive cancer. Gaining a better understanding of DCIS progression may reduce overtreatment of patients. Expression of the pro-inflammatory cytokine interleukin-6 increases with pathological stage and grade, and is associated with poorer prognosis in breast cancer patients. Carcinoma associated fibroblasts (CAFs), which are present in the stroma of DCIS patients are known to secrete pro-inflammatory cytokines and promote tumor progression. METHODS: We hypothesized that IL-6 paracrine signaling between DCIS cells and CAFs mediates DCIS proliferation and migration. To test this hypothesis, we utilized the mammary architecture and microenvironment engineering or MAME model to study the interactions between human breast CAFs and human DCIS cells in 3D over time. We specifically inhibited autocrine and paracrine IL-6 signaling to determine its contribution to early stage tumor progression. RESULTS: Here, DCIS cells formed multicellular structures that exhibited increased proliferation and migration when cultured with CAFs. Treatment with an IL-6 neutralizing antibody inhibited growth and migration of the multicellular structures. Moreover, selective knockdown of IL-6 in CAFs, but not in DCIS cells, abrogated the migratory phenotype. CONCLUSION: Our results suggest that paracrine IL-6 signaling between preinvasive DCIS cells and stromal CAFs represent an important factor in the initiation of DCIS progression to invasive breast carcinoma.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Fibroblastos/metabolismo , Interleucina-6/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Invasividad Neoplásica/patología
13.
Nat Rev Cancer ; 6(10): 764-75, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16990854

RESUMEN

Cysteine cathepsins are highly upregulated in a wide variety of cancers by mechanisms ranging from gene amplification to post-transcriptional modification. Their localization within intracellular lysosomes often changes during neoplastic progression, resulting in secretion of both inactive and active forms and association with binding partners on the tumour cell surface. Secreted, cell-surface and intracellular cysteine cathepsins function in proteolytic pathways that increase neoplastic progression. Direct proof for causal roles in tumour growth, migration, invasion, angiogenesis and metastasis has been shown by downregulating or ablating the expression of individual cysteine cathepsins in tumour cells and in transgenic mouse models of human cancer.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Neoplasias/enzimología , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
14.
J Physiol ; 592(23): 5251-68, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25260629

RESUMEN

In cystic fibrosis (CF) lung disease, the absence of functional CF transmembrane conductance regulator results in Cl(-)/HCO3 (-) hyposecretion and triggers Na(+) hyperabsorption through the epithelial Na(+) channel (ENaC), which contribute to reduced airway surface liquid (ASL) pH and volume. Prostasin, a membrane-anchored serine protease with trypsin-like substrate specificity has previously been shown to activate ENaC in CF airways. However, prostasin is typically inactive below pH 7.0, suggesting that it may be less relevant in acidic CF airways. Cathepsin B (CTSB) is present in both normal and CF epithelia and is secreted into ASL, but little is known about its function in the airways. We hypothesized that the acidic ASL seen in CF airways may stimulate CTSB to activate ENaC, contributing to Na(+) hyperabsorption and depletion of CF ASL volume. In Xenopus laevis oocytes, CTSB triggered α- and γENaC cleavage and induced an increase in ENaC activity. In bronchial epithelia from both normal and CF donor lungs, CTSB localized to the apical membrane. In normal and CF human bronchial epithelial cultures, CTSB was detected at the apical plasma membrane and in the ASL. CTSB activity was significantly elevated in acidic ASL, which correlated with increased abundance of ENaC in the plasma membrane and a reduction in ASL volume. This acid/CTSB-dependent activation of ENaC was ameliorated with the cell impermeable, CTSB-selective inhibitor CA074, suggesting that CTSB inhibition may have therapeutic relevance. Taken together, our data suggest that CTSB is a pathophysiologically relevant protease that activates ENaC in CF airways.


Asunto(s)
Catepsina B/metabolismo , Fibrosis Quística/metabolismo , Sodio/metabolismo , Animales , Catepsina B/antagonistas & inhibidores , Membrana Celular/metabolismo , Células Cultivadas , Quimotripsina/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Fibrosis Quística/tratamiento farmacológico , Dipéptidos/farmacología , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Femenino , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Oocitos/metabolismo , Subunidades de Proteína , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/metabolismo , Xenopus laevis
15.
Am J Pathol ; 182(2): 449-59, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23219716

RESUMEN

Hindgut-derived endoderm can differentiate into rectal, prostatic, and bladder phenotypes. Stromal-epithelial interactions are crucial for this development; however, the precise mechanisms by which epithelium responds to stromal cues remain unknown. We have previously reported ectopic expression of peroxisome proliferator-activated receptor-γ2 (PPARγ2) increased androgen receptor expression and promoted differentiation of mouse prostate epithelium. PPARγ is also implicated in urothelial differentiation. Herein we demonstrate that knockdown of PPARγ2 in benign human prostate epithelial cells (BHPrEs) promotes urothelial transdifferentiation. Furthermore, in vitro and in vivo heterotypic tissue regeneration models with embryonic bladder mesenchyme promoted urothelial differentiation of PPARγ2-deficient BHPrE cells, and deficiency of both PPARγ isoforms 1 and 2 arrested differentiation. Because PTEN deficiency is cooperative in urothelial pathogenesis, we engineered BHPrE cells with combined knockdown of PPARγ and PTEN and performed heterotypic recombination experiments using embryonic bladder mesenchyme. Whereas PTEN deficiency alone induced latent squamous differentiation in BHPrE cells, combined PPARγ and PTEN deficiency accelerated the development of keratinizing squamous metaplasia (KSM). We further confirmed via immunohistochemistry that gene expression changes in metaplastic recombinants reflected human urothelium undergoing KSM. In summary, these data suggest that PPARγ isoform expression provides a molecular basis for observations that adult human epithelium can be transdifferentiated on the basis of heterotypic mesenchymal induction. These data also implicate PPARγ and PTEN inactivation in the development of KSM.


Asunto(s)
Modelos Biológicos , PPAR gamma/deficiencia , Fosfohidrolasa PTEN/deficiencia , Regeneración , Urotelio/metabolismo , Urotelio/patología , Adulto , Animales , Secuencia de Bases , Línea Celular , Transdiferenciación Celular , Técnicas de Cocultivo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Hiperplasia , Mesodermo/metabolismo , Mesodermo/patología , Metaplasia , Ratones , Datos de Secuencia Molecular , PPAR gamma/metabolismo , Fosfohidrolasa PTEN/metabolismo , Urotelio/fisiopatología
16.
Biochim Biophys Acta ; 1824(1): 123-32, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21854877

RESUMEN

Our laboratory has had a longstanding interest in how the interactions between tumors and their microenvironment affect malignant progression. Recently, we have focused on defining the proteolytic pathways that function in the transition of breast cancer from the pre-invasive lesions of ductal carcinoma in situ (DCIS) to invasive ductal carcinomas (IDCs). We use live-cell imaging to visualize, localize and quantify proteolysis as it occurs in real-time and thereby have established roles for lysosomal cysteine proteases both pericellularly and intracellularly in tumor proteolysis. To facilitate these studies, we have developed and optimized 3D organotypic co-culture models that recapitulate the in vivo interactions of mammary epithelial cells or tumor cells with stromal and inflammatory cells. Here we will discuss the background that led to our present studies as well as the techniques and models that we employ. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.


Asunto(s)
Microambiente Celular/fisiología , Diagnóstico por Imagen/métodos , Neoplasias/metabolismo , Proteolisis , Microambiente Tumoral/fisiología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Células Cultivadas , Femenino , Humanos , Microscopía por Video , Modelos Biológicos , Neoplasias/diagnóstico , Neoplasias/patología , Análisis de la Célula Individual/métodos
17.
Angiogenesis ; 16(1): 223-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23096928

RESUMEN

Cathepsin B has been shown to be important in angiogenesis; therefore, understanding its regulation in endothelial cells should provide fundamental information that will aid in the development of new treatment options. Peroxisome proliferator-activated receptors (PPARs) have been shown to have anti-inflammatory, anti-angiogenic and anti-tumorigenic properties. We explored the influence of a PPARα agonist on cathepsin B expression in human endothelial cells. The PPARα agonist, Wy14643, was found to inhibit cathepsin B protein expression. Further studies demonstrated the Wy14643-dependent but PPARα-independent suppression of cathepsin B. This has been previously described for other PPAR agonists. Wy14643 suppressed the accumulation of cathepsin B mRNA, which was accompanied by the selective suppression of a 5'-alternative splice variant. Consistent with these results, luciferase promoter assays and electrophoretic mobility shift analysis demonstrated that the suppression was facilitated by reduced binding of the transcription factors USF1/2 to an E-box within the cathepsin B promoter. Additionally, Wy14643 treatment resulted in a reduction in cathepsin B half-life, suggesting a posttranslational regulatory mechanism. Overall, our results suggest that the PPARα-dependent anti-angiogenic action of Wy14643 seems to be mediated, in part, by Wy14643-dependent but PPARα-independent regulation of cathepsin B expression.


Asunto(s)
Catepsina B/genética , Catepsina B/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , PPAR alfa/agonistas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirimidinas/farmacología , Transcripción Genética/efectos de los fármacos , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Emparejamiento Base/genética , Western Blotting , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Elementos E-Box/genética , Exones/genética , Semivida , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , PPAR alfa/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Procesamiento Proteico-Postraduccional/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Factores Estimuladores hacia 5'/metabolismo
18.
Biol Chem ; 394(2): 307-16, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23152404

RESUMEN

Osteoarthritis and rheumatoid arthritis are destructive joint diseases that involve the loss of articular cartilage. Degradation of cartilage extracellular matrix is believed to occur due to imbalance between the catabolic and anabolic processes of resident chondrocytes. Previous work has suggested that various lysosomal cysteine cathepsins participate in cartilage degeneration; however, their exact roles in disease development and progression have not been elucidated. In order to study degradation processes under conditions resembling the in vivo milieu of the cartilage, we cultivated chondrocytes on a type II collagen-containing matrix. Stimulation of the cultivated chondrocytes with interleukin-1α and/or tumor necrosis factor α resulted in a time-dependent increase in cathepsin S expression and induced its secretion into the conditioned media. Using a novel bioluminescent activity-based probe, we were able to demonstrate a significant increase in proteolytic activity of cathepsin S in the conditioned media of proinflammatory cytokine-stimulated chondrocytes. For the first time, cathepsin S was demonstrated to be secreted from chondrocytes upon stimulation with the proinflammatory cytokines, and displayed proteolytic activity in culture supernatants. Its stability at neutral pH and potent proteolytic activity on extracellular matrix components mean that cathepsin S may contribute significantly to cartilage degradation and may thus be considered a potential drug target in joint diseases.


Asunto(s)
Catepsinas/biosíntesis , Catepsinas/metabolismo , Condrocitos/metabolismo , Inflamación/metabolismo , Interleucina-1alfa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Humanos , Proteolisis
19.
Proc Natl Acad Sci U S A ; 107(4): 1284-8, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20080563

RESUMEN

A chiral porphyrazine (pz), H(2)[pz(trans-A(2)B(2))] (247), has been prepared that exhibits preferential in vivo accumulation in the cells of tumors. Pz 247 exhibits near-infrared (NIR) emission with lambda > 700 nm in the required wavelength range for maximum tissue penetration. When MDA-MB-231 breast tumor cells are treated with 247, the agent shows strong intracellular fluorescence with an emission maximum, 704 nm, which indicates that it localizes within a hydrophobic microenvironment. Pz 247 is shown to associate with the lipophilic core of LDL and undergo cellular entry primarily through receptor-mediated endocytosis accumulating in lysosomes. Preliminary in vivo studies show that 247 exhibits preferential accumulation and retention in the cells of MDA-MB-231 tumors subcutaneously implanted in mice, thereby enabling NIR optical imaging with excellent contrast between tumor and surrounding tissue. The intensity of fluorescence from 247 within the tumor increases over time up to 48 h after injection presumably due to the sequestration of circulating 247/LDL complex by the tumor tissue. As the need for cholesterol, and thus LDL, is elevated in highly proliferative tumor cells over nontumorigenic cells, 247 has potential application for all such tumors.


Asunto(s)
Indoles/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Porfirinas/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Indoles/química , Ratones , Ratones SCID , Estructura Molecular , Porfirinas/química , Espectrometría de Fluorescencia , Espectroscopía Infrarroja Corta , Estereoisomerismo , Trasplante Heterólogo
20.
Biol Chem ; 393(12): 1405-16, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23667900

RESUMEN

The expression of the cysteine protease cathepsin B is increased in early stages of human breast cancer.To assess the potential role of cathepsin B in premalignant progression of breast epithelial cells, we employed a 3D reconstituted basement membrane overlay culture model of MCF10A human breast epithelial cells and isogenic variants that replicate the in vivo phenotypes of hyper plasia(MCF10AneoT) and atypical hyperplasia (MCF10AT1). MCF10A cells developed into polarized acinar structures with central lumens. In contrast, MCF10AneoT and MCF10AT1 cells form larger structures in which the lumens are filled with cells. CA074Me, a cell-permeable inhibitor selective for the cysteine cathepsins B and L,reduced proliferation and increased apoptosis of MCF10A, MCF10AneoT and MCF10AT1 cells in 3D culture. We detected active cysteine cathepsins in the isogenic MCF10 variants in 3D culture with GB111, a cell-permeable activity based probe, and established differential inhibition of cathepsin B in our 3D cultures. We conclude that cathepsin B promotes proliferation and premalignant progression of breast epithelial cells. These findings are consistent with studies by others showing that deletion of cathepsin B in the transgenic MMTV-PyMT mice, a murine model that is predisposed to development of mammary cancer, reduces malignant progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Mama/patología , Catepsina B/metabolismo , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/patología , Apoptosis/efectos de los fármacos , Mama/citología , Mama/efectos de los fármacos , Mama/enzimología , Neoplasias de la Mama/tratamiento farmacológico , Catepsina B/antagonistas & inhibidores , Catepsina L/antagonistas & inhibidores , Catepsina L/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Femenino , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA