Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Med ; 13(3): e6812, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38239047

RESUMO

BACKGROUND: It has been shown that tumor microenvironment (TME) hydroxyapatite (HAP) is typically associated with many malignancies and plays a role in tumor progression and growth. Additionally, acidosis in the TME has been reported to play a key role in selecting for a more aggressive tumor phenotype, drug resistance and desensitization to immunotherapy for many types of cancers. TME-HAP is an attractive target for tumor detection and treatment development since HAP is generally absent from normal soft tissue. We provide strong evidence that dissolution of hydroxyapatite (HAP) within the tumor microenvironment (TME-HAP) using a novel therapeutic can be used to kill cancer cells both in vitro and in vivo with minimal adverse effects. METHODS: We developed an injectable cation exchange nano particulate sulfonated polystyrene solution (NSPS) that we engineered to dissolve TME-HAP, inducing localized acute alkalosis and inhibition of tumor growth and glucose metabolism. This was evaluated in cell culture using 4T1, MDA-MB-231 triple negative breast cancer cells, MCF10 normal breast cells, and H292 lung cancer cells, and in vivo using orthotopic mouse models of cancer that contained detectable microenvironment HAP including breast (MMTV-Neu, 4T1, and MDA-MB-231), prostate (PC3) and colon (HCA7) cancer using 18 F-NaF for HAP and 18 F-FDG for glucose metabolism with PET imaging. On the other hand, H292 lung tumor cells that lacked detectable microenvironment HAP and MCF10a normal breast cells that do not produce HAP served as negative controls. Tumor microenvironment pH levels following injection of NSPS were evaluated via Chemical Exchange Saturation (CEST) MRI and via ex vivo methods. RESULTS: Within 24 h of adding the small concentration of 1X of NSPS (~7 µM), we observed significant tumor cell death (~ 10%, p < 0.05) in 4T1 and MDA-MB-231 cell cultures that contain HAP but ⟨2% in H292 and MCF10a cells that lack detectable HAP and in controls. Using CEST MRI, we found extracellular pH (pHe) in the 4T1 breast tumors, located in the mammary fat pad, to increase by nearly 10% from baseline before gradually receding back to baseline during the first hour post NSPS administration. in the tumors that contained TME-HAP in mouse models, MMTV-Neu, 4T1, and MDA-MB-231, PC3, and HCA7, there was a significant reduction (p<0.05) in 18 F-Na Fuptake post NSPS treatment as expected; 18 F- uptake in the tumor = 3.8 ± 0.5 %ID/g (percent of the injected dose per gram) at baseline compared to 1.8 ±0.5 %ID/g following one-time treatment with 100 mg/kg NSPS. Of similar importance, is that 18 F-FDG uptake in the tumors was reduced by more than 75% compared to baseline within 24 h of treatment with one-time NSPS which persisted for at least one week. Additionally, tumor growth was significantly slower (p < 0.05) in the mice treated with one-time NSPS. Toxicity showed no evidence of any adverse effects, a finding attributed to the absence of HAP in normal soft tissue and to our therapeutic NSPS having limited penetration to access HAP within skeletal bone. CONCLUSION: Dissolution of TME-HAP using our novel NSPS has the potential to provide a new treatment paradigm to enhance the management of cancer patients with poor prognosis.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Neoplasias Pulmonares , Humanos , Masculino , Animais , Camundongos , Preparações Farmacêuticas , Fluordesoxiglucose F18 , Imunoterapia , Alcanossulfonatos , Glucose , Hidroxiapatitas , Microambiente Tumoral
2.
Int J Mol Sci ; 20(20)2019 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-31652545

RESUMO

Inflammatory processes in the skin augment collagen degradation due to the up-regulation of matrix metalloproteinases (MMPs). The aim of the present project was to study the specific impact of MMP-3 on collagen loss in skin and its interplay with the collagenase MMP-13 under inflammatory conditions mimicked by the addition of the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α). Skin explants from MMP-3 knock-out (KO) mice or from transgenic (TG) mice overexpressing MMP-3 in the skin and their respective wild-type counterparts (WT and WTT) were incubated ex vivo for eight days. The rate of collagen degradation, measured by released hydroxyproline, was reduced (p < 0.001) in KO skin explants compared to WT control skin but did not differ (p = 0.47) between TG and WTT skin. Treatment with the MMP inhibitor GM6001 reduced hydroxyproline media levels from WT, WTT and TG but not from KO skin explants. TNF-α increased collagen degradation in the WT group (p = 0.0001) only. More of the active form of MMP-13 was observed in the three MMP-3 expressing groups (co-incubation with receptor-associated protein stabilized MMP-13 subforms and enhanced detection in the media). In summary, the innate level of MMP-3 seems responsible for the accelerated loss of cutaneous collagen under inflammatory conditions, possibly via MMP-13 in mice.


Assuntos
Colágeno/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Pele/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Dipeptídeos/farmacologia , Masculino , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/genética , Inibidores de Metaloproteinases de Matriz/farmacologia , Camundongos , Proteólise , Pele/efeitos dos fármacos
3.
Lab Chip ; 19(5): 864-874, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30720811

RESUMO

Microfluidic organ-on-chip devices constructed from polydimethylsiloxane (PDMS) have proven useful in studying both beneficial and adverse effects of drugs, supplements, and potential toxicants. Despite multiple advantages, one clear drawback of PDMS-based devices is binding of hydrophobic chemicals to their exposed surfaces. Chemical binding to PDMS can change the timing and extent of chemical delivery to cells in such devices, potentially altering dose-response curves. Recent efforts have quantified PDMS binding for selected chemicals. Here, we test a wider set of nineteen chemicals using UV-vis or infrared spectroscopy to characterize loss of chemical from solution in two setups with different PDMS-surface-to-solution-volume ratios. We find discernible PDMS binding for eight chemicals and show that PDMS binding is strongest for chemicals with a high octanol-water partition coefficient (log P > 1.85) and low H-bond donor number. Further, by measuring depletion and return of chemical from solution over tens to hundreds of hours and fitting these results to a first order model of binding kinetics, we characterize partitioning into PDMS in terms of binding capacities per unit surface area and both forward and reverse rate constants. These fitted parameters were used to model the impact of PDMS binding on chemical transport and bioavailability under realistic flow conditions and device geometry. The models predict that PDMS binding could alter in-device cellular exposures for both continuous and bolus dosing schemes by up to an order of magnitude compared to nominal input doses.


Assuntos
Dimetilpolisiloxanos/química , Dimetilpolisiloxanos/farmacocinética , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas , Sítios de Ligação , Disponibilidade Biológica , Dimetilpolisiloxanos/síntese química , Cinética , Estrutura Molecular
4.
J R Soc Interface ; 15(138)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29367239

RESUMO

A main goal of mathematical and computational oncology is to develop quantitative tools to determine the most effective therapies for each individual patient. This involves predicting the right drug to be administered at the right time and at the right dose. Such an approach is known as precision medicine. Mathematical modelling can play an invaluable role in the development of such therapeutic strategies, since it allows for relatively fast, efficient and inexpensive simulations of a large number of treatment schedules in order to find the most effective. This review is a survey of mathematical models that explicitly take into account the spatial architecture of three-dimensional tumours and address tumour development, progression and response to treatments. In particular, we discuss models of epithelial acini, multicellular spheroids, normal and tumour spheroids and organoids, and multi-component tissues. Our intent is to showcase how these in silico models can be applied to patient-specific data to assess which therapeutic strategies will be the most efficient. We also present the concept of virtual clinical trials that integrate standard-of-care patient data, medical imaging, organ-on-chip experiments and computational models to determine personalized medical treatment strategies.


Assuntos
Biologia Computacional/métodos , Modelos Biológicos , Neoplasias , Medicina de Precisão/métodos , Esferoides Celulares , Humanos , Oncologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Células Tumorais Cultivadas
5.
J Neuroinflammation ; 13(1): 306, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27955696

RESUMO

BACKGROUND: Understanding blood-brain barrier responses to inflammatory stimulation (such as lipopolysaccharide mimicking a systemic infection or a cytokine cocktail that could be the result of local or systemic inflammation) is essential to understanding the effect of inflammatory stimulation on the brain. It is through the filter of the blood-brain barrier that the brain responds to outside influences, and the blood-brain barrier is a critical point of failure in neuroinflammation. It is important to note that this interaction is not a static response, but one that evolves over time. While current models have provided invaluable information regarding the interaction between cytokine stimulation, the blood-brain barrier, and the brain, these approaches-whether in vivo or in vitro-have often been only snapshots of this complex web of interactions. METHODS: We utilize new advances in microfluidics, organs-on-chips, and metabolomics to examine the complex relationship of inflammation and its effects on blood-brain barrier function ex vivo and the metabolic consequences of these responses and repair mechanisms. In this study, we pair a novel dual-chamber, organ-on-chip microfluidic device, the NeuroVascular Unit, with small-volume cytokine detection and mass spectrometry analysis to investigate how the blood-brain barrier responds to two different but overlapping drivers of neuroinflammation, lipopolysaccharide and a cytokine cocktail of IL-1ß, TNF-α, and MCP1,2. RESULTS: In this study, we show that (1) during initial exposure to lipopolysaccharide, the blood-brain barrier is compromised as expected, with increased diffusion and reduced presence of tight junctions, but that over time, the barrier is capable of at least partial recovery; (2) a cytokine cocktail also contributes to a loss of barrier function; (3) from this time-dependent cytokine activation, metabolic signature profiles can be obtained for both the brain and vascular sides of the blood-brain barrier model; and (4) collectively, we can use metabolite analysis to identify critical pathways in inflammatory response. CONCLUSIONS: Taken together, these findings present new data that allow us to study the initial effects of inflammatory stimulation on blood-brain barrier disruption, cytokine activation, and metabolic pathway changes that drive the response and recovery of the barrier during continued inflammatory exposure.


Assuntos
Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Citocinas/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Claudina-5/metabolismo , Citocinas/farmacologia , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/farmacologia , Dispositivos Lab-On-A-Chip , Lipopolissacarídeos/farmacologia , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/genética , Modelos Biológicos , Transporte Proteico/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/farmacologia , Proteína da Zônula de Oclusão-1/metabolismo
6.
Biomicrofluidics ; 9(5): 054124, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26576206

RESUMO

The blood-brain barrier (BBB) is a critical structure that serves as the gatekeeper between the central nervous system and the rest of the body. It is the responsibility of the BBB to facilitate the entry of required nutrients into the brain and to exclude potentially harmful compounds; however, this complex structure has remained difficult to model faithfully in vitro. Accurate in vitro models are necessary for understanding how the BBB forms and functions, as well as for evaluating drug and toxin penetration across the barrier. Many previous models have failed to support all the cell types involved in the BBB formation and/or lacked the flow-created shear forces needed for mature tight junction formation. To address these issues and to help establish a more faithful in vitro model of the BBB, we have designed and fabricated a microfluidic device that is comprised of both a vascular chamber and a brain chamber separated by a porous membrane. This design allows for cell-to-cell communication between endothelial cells, astrocytes, and pericytes and independent perfusion of both compartments separated by the membrane. This NeuroVascular Unit (NVU) represents approximately one-millionth of the human brain, and hence, has sufficient cell mass to support a breadth of analytical measurements. The NVU has been validated with both fluorescein isothiocyanate (FITC)-dextran diffusion and transendothelial electrical resistance. The NVU has enabled in vitro modeling of the BBB using all human cell types and sampling effluent from both sides of the barrier.

7.
Carcinogenesis ; 36 Suppl 1: S128-59, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26106135

RESUMO

The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.


Assuntos
Carcinógenos Ambientais/efeitos adversos , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Animais , Progressão da Doença , Exposição Ambiental/efeitos adversos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos
8.
Biomed Microdevices ; 16(1): 91-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24065585

RESUMO

Polydimethylsiloxane (PDMS) is a commonly used polymer in the fabrication of microfluidic devices due to such features as transparency, gas permeability, and ease of patterning with soft lithography. The surface characteristics of PDMS can also be easily changed with oxygen or low pressure air plasma converting it from a hydrophobic to a hydrophilic state. As part of such a transformation, surface methyl groups are removed and replaced with hydroxyl groups making the exposed surface to resemble silica, a gas impermeable substance. We have utilized Platinum(II)-tetrakis(pentaflourophenyl)porphyrin immobilized within a thin (~1.5 um thick) polystyrene matrix as an oxygen sensor, Stern-Volmer relationship, and Fick's Law of simple diffusion to measure the effects of PDMS composition, treatment, and storage on oxygen diffusion through PDMS. Results indicate that freshly oxidized PDMS showed a significantly smaller diffusion coefficient, indicating that the SiO2 layer formed on the PDMS surface created an impeding barrier. This barrier disappeared after a 3-day storage in air, but remained significant for up to 3 weeks if PDMS was maintained in contact with water. Additionally, higher density PDMS formulation (5:1 ratio) showed similar diffusion characteristics as normal (10:1 ratio) formulation, but showed 60 % smaller diffusion coefficient after plasma treatment that never recovered to pre-treatment levels even after a 3-week storage in air. Understanding how plasma surface treatments contribute to oxygen diffusion will be useful in exploiting the gas permeability of PDMS to establish defined normoxic and hypoxic oxygen conditions within microfluidic bioreactor systems.


Assuntos
Dimetilpolisiloxanos/química , Gases/química , Técnicas de Cultura de Células , Difusão , Técnicas Analíticas Microfluídicas/instrumentação , Microfluídica/métodos , Oxirredução , Oxigênio/química , Permeabilidade , Poliestirenos/química , Dióxido de Silício/química , Propriedades de Superfície , Água/química
9.
Stem Cell Res Ther ; 4 Suppl 1: S18, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24564885

RESUMO

The blood-brain barrier (BBB) dynamically controls exchange between the brain and the body, but this interaction cannot be studied directly in the intact human brain or sufficiently represented by animal models. Most existing in vitro BBB models do not include neurons and glia with other BBB elements and do not adequately predict drug efficacy and toxicity. Under the National Institutes of Health Microtissue Initiative, we are developing a three-dimensional, multicompartment, organotypic microphysiological system representative of a neurovascular unit of the brain. The neurovascular unit system will serve as a model to study interactions between the central nervous system neurons and the cerebral spinal fluid (CSF) compartment, all coupled to a realistic blood-surrogate supply and venous return system that also incorporates circulating immune cells and the choroid plexus. Hence all three critical brain barriers will be recapitulated: blood-brain, brain-CSF, and blood-CSF. Primary and stem cell-derived human cells will interact with a variety of agents to produce critical chemical communications across the BBB and between brain regions. Cytomegalovirus, a common herpesvirus, will be used as an initial model of infections regulated by the BBB. This novel technological platform, which combines innovative microfluidics, cell culture, analytical instruments, bioinformatics, control theory, neuroscience, and drug discovery, will replicate chemical communication, molecular trafficking, and inflammation in the brain. The platform will enable targeted and clinically relevant nutritional and pharmacologic interventions for or prevention of such chronic diseases as obesity and acute injury such as stroke, and will uncover potential adverse effects of drugs. If successful, this project will produce clinically useful technologies and reveal new insights into how the brain receives, modifies, and is affected by drugs, other neurotropic agents, and diseases.


Assuntos
Encéfalo/metabolismo , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/citologia , Líquido Cefalorraquidiano/fisiologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/toxicidade
10.
Lab Chip ; 12(21): 4560-8, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-22964798

RESUMO

We have developed a novel, portable, gravity-fed, microfluidics-based platform suitable for optical interrogation of long-term organotypic cell culture. This system is designed to provide convenient control of cell maintenance, nutrients, and experimental reagent delivery to tissue-like cell densities housed in a transparent, low-volume microenvironment. To demonstrate the ability of our Thick-Tissue Bioreactor (TTB) to provide stable, long-term maintenance of high-density cellular arrays, we observed the morphogenic growth of human mammary epithelial cell lines, MCF-10A and their invasive variants, cultured under three-dimensional (3D) conditions inside our system. Over the course of 21 days, these cells typically develop into hollow "mammospheres" if cultured in standard 3D Matrigel. This complex morphogenic process requires alterations in a variety of cellular functions, including degradation of extracellular matrix that is regulated by cell-produced matrix proteinases. For our "drug" delivery testing and validation experiments we have introduced proteinase inhibitors into the fluid supply system, and we observed both reduced proteinase activity and inhibited cellular morphogenesis. The size inhibition results correlated well with the overall proteinase activities of the tested cells.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/métodos , Sistemas de Liberação de Medicamentos , Células Epiteliais/efeitos dos fármacos , Técnicas Analíticas Microfluídicas/métodos , Inibidores de Proteases/farmacologia , Técnicas de Cultura de Células/instrumentação , Células Cultivadas , Docetaxel , Células Epiteliais/enzimologia , Desenho de Equipamento , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Peptídeo Hidrolases/metabolismo , Relação Estrutura-Atividade , Taxoides/farmacologia
11.
Biomed Microdevices ; 13(5): 837-46, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21710371

RESUMO

Morphogenesis is a fundamental process by which new blood vessels are formed during angiogenesis. The ability to control angiogenesis would lead to improvements in tissue engineering constructions; indeed, the study of angiogenesis has numerous clinical applications, for example, in the investigation of metastatic cancer, peripheral and coronary vascular disease, and wound healing. Conventional in vitro organotypic cell culture approaches to these studies are limited primarily by their reliance on microvascular vessel formation through a random process of morphogenesis that lacks the spatial reproducibility and orientation needed for high-throughput drug testing. We have developed a bioreactor system for scaffold-guided tubulogenesis coupled with 3-D organotypic culture to spatially control vessel formation and its orientation. To create microchannels to guide microvessel formation, we fabricated rigid scaffolds using photolithography and light curing epoxy, and soft scaffolds formed by a polydimethylsiloxane (PDMS) stamp directly into collagen. Scaffolds seeded with dermal microvascular endothelial cells were placed between gelled layers of collagen containing dermal fibroblasts within a Transwell filter system and cultured for up to 2 weeks to allow for vessel maturation. Morphological analysis of thin tissue sections following standard histology and immunohistochemical detection of endothelial cells, fibroblasts, and basement membrane confirmed vessel formation along the microchannel walls with either scaffold. This system may also provide a means to explore revascularization within decellularized extracellular matrices, the culture of microvessel networks with controlled geometries, and possibly the spatial guidance of angiogenesis for interfacing with an external microfluidic supply network. As a new tool for guided angiogenesis, our approach introduces new possibilities for identification of anti-angiogenic therapeutics.


Assuntos
Endotélio Vascular/citologia , Microvasos/crescimento & desenvolvimento , Alicerces Teciduais , Membrana Basal , Reatores Biológicos , Técnicas de Cultura de Células/métodos , Colágeno/química , Dimetilpolisiloxanos/química , Fibroblastos , Humanos , Microvasos/citologia , Morfogênese , Reprodutibilidade dos Testes , Propriedades de Superfície
12.
Exp Biol Med (Maywood) ; 235(4): 411-23, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20407073

RESUMO

In its simplest description, a tumor is comprised of an expanding population of transformed cells supported by a surrounding microenvironment termed the tumor stroma. The tumor microenvironment has a very complex composition, including multiple types of stromal cells, a dense network of various extracellular matrix (ECM) fibers interpenetrated by the interstitial fluid and gradients of several chemical species that either are dissolved in the fluid or are bound to the ECM structure. In order to study experimentally such complex interactions between multiple players, cancer is dissected and considered at different scales of complexity, such as protein interactions, biochemical pathways, cellular functions or whole organism studies. However, the integration of information acquired from these studies into a common description is as difficult as the disease itself. Computational models of cancer can provide cancer researchers with invaluable tools that are capable of integrating the complexity into organizing principles as well as suggesting testable hypotheses. We will focus in this Minireview on mathematical models in which the whole cell is a main modeling unit. We will present a current stage of such cell-focused mathematical modeling incorporating different stromal components and their interactions with growing tumors, and discuss what modeling approaches can be undertaken to complement the in vivo and in vitro experimentation.


Assuntos
Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Ensaios Clínicos como Assunto , Simulação por Computador , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Células Estromais/citologia , Células Estromais/metabolismo , Células Estromais/patologia
13.
Math Biosci Eng ; 6(3): 521-46, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19566124

RESUMO

In this paper we consider chemotherapy in a spatial model of tumor growth. The model, which is of reaction-diffusion type, takes into account the complex interactions between the tumor and surrounding stromal cells by including densities of endothelial cells and the extra-cellular matrix. When no treatment is applied the model reproduces the typical dynamics of early tumor growth. The initially avascular tumor reaches a diffusion limited size of the order of millimeters and initiates angiogenesis through the release of vascular endothelial growth factor (VEGF) secreted by hypoxic cells in the core of the tumor. This stimulates endothelial cells to migrate towards the tumor and establishes a nutrient supply sufficient for sustained invasion. To this model we apply cytostatic treatment in the form of a VEGF-inhibitor, which reduces the proliferation and chemotaxis of endothelial cells. This treatment has the capability to reduce tumor mass, but more importantly, we were able to determine that inhibition of endothelial cell proliferation is the more important of the two cellular functions targeted by the drug. Further, we considered the application of a cytotoxic drug that targets proliferating tumor cells. The drug was treated as a diffusible substance entering the tissue from the blood vessels. Our results show that depending on the characteristics of the drug it can either reduce the tumor mass significantly or in fact accelerate the growth rate of the tumor. This result seems to be due to complicated interplay between the stromal and tumor cell types and highlights the importance of considering chemotherapy in a spatial context.


Assuntos
Antineoplásicos/farmacologia , Modelos Imunológicos , Neoplasias/imunologia , Neovascularização Patológica/imunologia , Taxoides/farmacologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Antineoplásicos/uso terapêutico , Simulação por Computador , Docetaxel , Células Endoteliais/imunologia , Humanos , Neoplasias/tratamento farmacológico , Taxoides/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
14.
Am J Pathol ; 173(5): 1528-39, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18832569

RESUMO

Matrix metalloproteinase (MMP)-3 is induced by multiple cell types in the skin during processes involved in both normal and pathological tissue remodeling. We previously demonstrated that MMP3-null animals have an increased sensitivity to the development of squamous cell carcinoma, suggesting that overall, MMP3 has a protective role in squamous cell carcinoma. However, not all cellular responses affected by a loss of MMP3 are tumor-protective, and tumor expression of MMP3 is co-incident with an invasive tumor phenotype. Transgenic mice were generated with MMP3 targeted to keratinocytes to examine the biological role of tumor-produced MMP3. Overexpression of MMP3 reduced tumor multiplicity in response to chemically induced squamous cell carcinoma. Vascular density was increased with MMP3 overexpression; however, other cellular processes, including tumor growth and leukocyte infiltration, were unaffected. In accordance with the change in tumor multiplicity, SP-1 murine papilloma cell lines that were generated to stably express MMP3 lost the capacity to establish palpable tumors following orthotopic injection into immunocompromised mice. Analysis of epidermal biopsies taken at 1 to 2 weeks postinjection revealed that these MMP3-expressing Sp-1 lines had reduced levels of proliferation and pronounced differentiation. These same cells demonstrated an increased ability to differentiate in vitro, an effect that was inhibited by broad-spectrum MMP and selective MMP3 inhibition. These studies suggest that keratinocyte expression of MMP3 promotes cellular differentiation, impeding tumor establishment during tumorigenesis.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/patologia , Diferenciação Celular , Queratinócitos/enzimologia , Queratinócitos/patologia , Metaloproteinase 3 da Matriz/metabolismo , Animais , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/induzido quimicamente , Bovinos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Modelos Animais de Doenças , Epiderme/enzimologia , Epiderme/patologia , Feminino , Marcação de Genes , Queratina-5/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neutrófilos/patologia , Ratos , Células Estromais/patologia , Transplante Homólogo
15.
Exp Cell Res ; 303(2): 308-20, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15652345

RESUMO

Matrix metalloproteinase-7 (MMP-7) is primarily expressed in glandular epithelium. Therefore, its mechanism of action may be influenced by its regulated vectorial release to either the apical and/or basolateral compartments, where it would act on its various substrates. To gain a better understanding of where MMP-7 is released in polarized epithelium, we have analyzed its pattern of secretion in polarized MDCK cells expressing stably transfected human MMP-7 (MDCK-MMP-7), and HCA-7 and Caco2 human colon cancer cell lines. In all cell lines, latent MMP-7 was secreted to both cellular compartments, but was 1.5- to 3-fold more abundant in the basolateral compartment as compared to the apical. However, studies in the MDCK system demonstrated that MMP-7 activity was 2-fold greater in the apical compartment of MDCK-MMP-7(HIGH)-polarized monolayers, which suggests the apical co-release of an MMP-7 activator. In functional assays, MMP-7 over-expression increased cell saturation density as a result of increased cell proliferation with no effect on apoptosis. Apical MMP-7 activity was shown to be responsible for the proliferative effect, which occurred, as demonstrated by media transfer experiments, through cleavage of an apical substrate and not through the generation of a soluble factor. Taken together, our findings demonstrate the importance of MMP-7 secretion in relation to its mechanism of action when expressed in a polarized epithelium.


Assuntos
Metaloendopeptidases/fisiologia , Animais , Células CACO-2 , Linhagem Celular , Membrana Celular/enzimologia , Polaridade Celular , Proliferação de Células , Cães , Humanos , Metaloproteinase 7 da Matriz , Metaloendopeptidases/genética , Transfecção
16.
Cancer Res ; 64(19): 6965-72, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15466188

RESUMO

Elevated expression of matrix metalloproteinase-3 (MMP-3/stromelysin-1) is associated with a variety of tumor types, although its in vivo functional role remains unclear. In human and murine squamous cell carcinoma (SCC), MMP-3 is expressed in the stromal compartment at all of the stages of tumor progression and is expressed by the malignant epithelial cells in late-stage, highly invasive tumors. To elucidate whether MMP-3 plays a causal role during SCC, wild-type and MMP-3 null mice were subjected to chemical carcinogenesis procedures by topical application of either the complete carcinogen 1-methyl-3-nitro-1-nitroso-guanidine or two-stage initiation and promotion with 7,12-dimethylbenz[a]anthracene and 12-O-tetradecanoylphorbol-13-acetate. Contrasting with our expectations, tumors originating on MMP-3 null mice had enhanced initial tumor growth rates as compared with control animals, although there was no difference in tumor onset or incidence. This elevated rate in growth was coupled with an elevated proliferative index and a reduced vasculature density but with no significant effect on apoptosis. Tumors from MMP-3 null mice had a prevalence of undifferentiated spindle tumors as compared with controls, which was concomitant with a higher percentage of MMP-3 null mice evidencing surface lung metastases. Tumor progression in MMP-3 null mice was inversely associated with leukocyte infiltration, in which an overall reduction in tumor-associated macrophages and neutrophils was evident. We propose that MMP-3 is expressed as a protective response and plays an important role in host defense during SCC tumorigenesis.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Metaloproteinase 3 da Matriz/fisiologia , Animais , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Divisão Celular/fisiologia , Progressão da Doença , Feminino , Humanos , Imunoquímica , Leucócitos/imunologia , Leucócitos/patologia , Masculino , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Células Estromais/enzimologia , Células Estromais/patologia
17.
Int J Oncol ; 25(2): 319-24, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254728

RESUMO

Fetuin, a major serum glycoprotein secreted by the liver, has been shown to play a role in bone development, calcium homeostasis and insulin sensitivity. In an earlier study, we demonstrated that bovine fetuin can bind to the plasma membrane of squamous and spindle-cell carcinoma cells. To test our hypothesis that fetuin plays a causal role in skin tumorigenesis, fetuin-A null and wild-type mice were challenged using a two-stage chemically-induced carcinogenesis protocol with DMBA (7,12-dimethylbenzo(a)anthracene) as the initiator, followed by twice weekly treatments with the tumor promoter TPA (12-O-tetradecanoylphorbol-13-acetate). Tumors that developed on fetuin-A null animals grew at a similar rate as those arising on their wild-type counterparts. Absence of fetuin-A did not alter tumor onset or conversion to squamous cell carcinoma, but reduced the number of tumors per mouse by 30%. This correlated with a decrease in tumor burden in fetuin-A null animals compared to wild-type weeks 18-22 from tumor onset. In addition, tumors arising on fetuin-A null mice had a diminished proliferative index with no change in cell survival or neovascularization in comparison with wild-type tumors. Our results suggest that fetuin-A contributes to early stages of skin tumorigenesis.


Assuntos
Proteínas Sanguíneas/fisiologia , Neoplasias Cutâneas/etiologia , Acetato de Tetradecanoilforbol/análogos & derivados , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Apoptose , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Pele/efeitos dos fármacos , Pele/patologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/farmacologia , alfa-2-Glicoproteína-HS
18.
Int J Oncol ; 21(5): 965-71, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12370742

RESUMO

Alpha2-HS glycoprotein (fetuin) is a major plasma glycoprotein predominantly synthesized by the liver. We have previously demonstrated that human fetuin produced by hepatocellular carcinoma cells can activate the matrix metalloproteinase MMP-9 (gelatinase-B). Stromelysin-1 (MMP-3) is over-expressed in murine skin tumors and is associated with a metastatic cell phenotype. We hypothesize that fetuin plays a role in tumor progression of cell types which by themselves do not have the ability to express fetuin. Immunohistochemical staining revealed that fetuin surrounds the tumor cells in murine squamous cell carcinoma tumor specimens, similar to the expression pattern previously seen for MMP-3. The physical association of fetuin and MMP-3 was demonstrated by immunoprecipitation of radiolabeled fetuin by antibodies to MMP-3. Fetuin facilitates the conversion of pro-MMP-3 to its active form, although this effect is indirect. The association of iodinated fetuin to the cell surface of intact cultured cells derived from a murine tumor with squamous (B9) and spindle (A5) morphologies was determined by binding experiments and Scatchard analysis. Fetuin binds with Bmax values in the range of 1.26-2.1 (mean = 1.7) fmol/1 x 10(5) cells for A5 cells, and 1.5-1.7 (mean = 1.6) fmol/1 x 10(5) cells for B9 cells. The mean KD was 0.46+/-0.19 nmol for both A5 and B9 cells. Our data therefore are consistent with the model that fetuin binds to the cell surface of tumor cells and acts to localize and anchor other molecules important during tumor progression to the plasma membrane.


Assuntos
Carcinoma de Células Escamosas/etiologia , Metaloproteinase 3 da Matriz/fisiologia , alfa-Fetoproteínas/fisiologia , Animais , Células CHO , Carcinoma de Células Escamosas/metabolismo , Cricetinae , Metaloproteinase 3 da Matriz/análise , Camundongos , Células Tumorais Cultivadas , alfa-Fetoproteínas/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...