Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
World J Surg ; 48(8): 1902-1911, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38890767

RESUMO

BACKGROUND: Patients undergoing major oncological abdominal surgery are prone to postoperative complications, making early recognition crucial. Clinical deterioration is often preceded by changes in vital signs, which are typically measured thrice a day by a nurse. However, intermittent measurements may delay recognizing clinical deterioration. Continuous vital parameter monitoring may lead to earlier recognition and management of complications and reduce nursing workload. OBJECTIVE: To compare vital parameter measurements between ward nurses and a wireless continuous monitoring system (Sensium® wireless patch) and assess whether this patch can detect clinical deterioration earlier in patients with complications in the first postoperative week. METHODS: Vital parameters (heart rate, respiratory rate, and temperature) were collected in patients undergoing an oncological resection of the liver, colorectal, or pancreas. Sensium® patch measurements were compared to nurses' measurements to assess the percentages of discordant measurements. In patients with complications in the first postoperative week, time discrepancies between nurses and Sensium® patch measurements were identified in cases of clinical deterioration (respiratory rate ≥15/min, heart rate ≥100/min, and temperature ≥38°C). RESULTS: Among 227 patients, 22% of the patients experienced complications. Nurse and Sensium® measurements were discrepant in 586/2272 measurements (26%). In 506/586 discrepancies (86%), this was due to the respiratory rate (difference ≥4/min). Compared to nurses, the Sensium® patch detected an elevated respiratory rate 14 h earlier and heart rate 2 h earlier within complications in the first postoperative week. For temperature, no difference was observed. CONCLUSION: Continuous monitoring with the Sensium® wireless patch holds promise for earlier recognition of complications in patients who underwent major oncological abdominal surgery.


Assuntos
Complicações Pós-Operatórias , Sinais Vitais , Humanos , Feminino , Masculino , Monitorização Fisiológica/métodos , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/epidemiologia , Idoso , Pessoa de Meia-Idade , Procedimentos Cirúrgicos do Sistema Digestório/efeitos adversos , Procedimentos Cirúrgicos do Sistema Digestório/métodos , Diagnóstico Precoce , Estudos Prospectivos , Tecnologia sem Fio
2.
Gastroenterology ; 149(3): 692-704, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25962936

RESUMO

BACKGROUND & AIMS: Colon tumors contain a fraction of undifferentiated stem cell-like cancer cells with high tumorigenic potential. Little is known about the signals that maintain these stem-like cells. We investigated whether differentiated tumor cells provide support. METHODS: We established undifferentiated colonosphere cultures from human colon tumors and used them to generate stably differentiated cell lines. Antibody arrays were used to identify secreted factors. Expression of genes involved in stemness, differentiation, and the epithelial to mesenchymal transition was measured using reverse transcription quantitative polymerase chain reaction. Expression of KIT in human tumors was analyzed with gene expression arrays and by immunohistochemistry. Colonospheres were injected into the livers of CBy.Cg-Foxn1nu/J mice. After liver tumors had formed, hypoxia was induced by vascular clamping. RESULTS: Differentiated cells from various tumors, or medium conditioned by them, increased the clonogenic capacity of colonospheres. Stem cell factor (SCF) was secreted by differentiated tumor cells and supported the clonogenic capacity of KIT(+) colonosphere cells. Differentiated tumor cells induced the epithelial to mesenchymal transition in colonosperes; this was prevented by inhibition of KIT or SCF. SCF prevented loss of clonogenic potential under differentiation-inducing conditions. Suppression of SCF or KIT signaling greatly reduced the expression of genes that regulate stemness and the epithelial to mesenchymal transition and inhibited clonogenicity and tumor initiation. Bioinformatic and immunohistochemical analyses revealed a correlation between expression of KIT- and hypoxia-related genes in colon tumors, which was highest in relapse-prone mesenchymal-type tumors. Hypoxia induced expression of KIT in cultured cells and in human colon tumor xenografts and this contributed to the clonogenic capacity of the tumor cells. CONCLUSIONS: Paracrine signaling from SCF to KIT, between differentiated tumor cells and undifferentiated stem-like tumor cells, helps maintain the stem-like features of tumor cells, predominantly under conditions of hypoxia.


Assuntos
Biomarcadores Tumorais/metabolismo , Diferenciação Celular , Neoplasias do Colo/enzimologia , Células-Tronco Neoplásicas/enzimologia , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/metabolismo , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Hipóxia Celular , Proliferação de Células , Técnicas de Cocultura , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Comunicação Parácrina/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais , Esferoides Celulares , Fator de Células-Tronco/antagonistas & inibidores , Fator de Células-Tronco/genética , Fatores de Tempo , Carga Tumoral , Células Tumorais Cultivadas
3.
Surgeon ; 13(2): 77-82, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24486032

RESUMO

BACKGROUND: Resection of colorectal liver metastases (CRLM) is often hindered by their location close to the major hepatic vessels. So far, radiofrequency ablation for perivascular tumours was thought to be ineffective and unsafe due to either the heat sink effect or vascular thrombosis. The aim of this study was to examine whether RFA using multipolar probes could be a safe and effective option for CRLM adjacent to major hepatic vessels. METHODS: Patients were treated with multipolar RFA during an open procedure using 3 simultaneously placed electrodes. In 52 consecutive patients with CRLM, 144 tumours were ablated with RFA. In 16 out of 52 (31%) patients, metastases were abutting major hepatic vessels. We examined whether perivascular location was a risk factor for local tumour progression. The relation between perivascular location and time to local tumour progression and recurrence free survival was assessed using cox-regression analysis. RESULTS: All patients were followed for at least 3 years after RFA unless they deceased before this time. Local tumour progression following RFA occurred in 17 out of 144 tumours (12%), of which 4 out of 21 were perivascular tumours. Tumour size was the only risk factor for local tumour progression in this study. Proximity to large vessels was neither a risk factor for local local tumour progression, nor for time to local tumour progression or recurrence free survival. DISCUSSION: This study indicates that patients with CRLM abutting any of the large hepatic vessels can be safe and effectively treated with RFA when using a multipolar system.


Assuntos
Ablação por Cateter , Neoplasias Colorretais/patologia , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/cirurgia , Fígado/irrigação sanguínea , Fígado/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Ablação por Cateter/instrumentação , Feminino , Artéria Hepática/cirurgia , Veias Hepáticas/cirurgia , Humanos , Neoplasias Hepáticas/secundário , Masculino , Pessoa de Meia-Idade , Veia Porta/cirurgia , Estudos Retrospectivos
4.
Ann Surg ; 259(4): 750-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24253142

RESUMO

OBJECTIVE: To assess the contribution of hypoxia and bone marrow-derived cells to aggressive outgrowth of micrometastases after liver surgery. BACKGROUND: Liver surgery generates a microenvironment that fosters aggressive tumor recurrence. These areas are characterized by chronic hypoxia and influx of bone marrow-derived cells. METHODS: The contribution of hematopoietic cell types was studied in mice lacking specific components of the immune system and in irradiated mice lacking all bone marrow-derived cells. Tumor cells were derived from colorectal cancer patients and from a metastatic tumor cell line. Hypoxia-induced changes in stem cell and differentiation marker expression, clone-forming potential, and metastatic capacity were assessed. The effect of vascular clamping on cancer stem cell (CSC) characteristics was performed in mice bearing patient-derived liver metastases. RESULTS: Immune cells and bone marrow-derived cells were not required for aggressive outgrowth of micrometastases in livers treated with surgery. Rather, hypoxia was sufficient to promote invasion and accelerate metastatic outgrowth. This was associated with a rapid loss of differentiation markers and increased expression of CSC markers and clone-forming capacity. Likewise, metastases residing in ischemia-reperfusion-injured liver lobes acquired CSC characteristics. Despite their renowned general resistance to chemotherapy, clone-forming CSCs were readily killed by the hypoxia-activated prodrug tirapazamine. CONCLUSIONS: Surgery-generated hypoxia in the liver causes rapid dedifferentiation of tumor cells into immature CSCs with high clone- and metastasis-forming capacity. The results help explain the phenomenon of aggressive local tumor recurrence after liver surgery and offer a potential strategy to kill aggressive CSCs by hypoxia-activated prodrugs.


Assuntos
Neoplasias Colorretais/patologia , Hepatectomia , Hipóxia/etiologia , Neoplasias Hepáticas Experimentais/secundário , Recidiva Local de Neoplasia/patologia , Neoplasia Residual/patologia , Complicações Pós-Operatórias , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Western Blotting , Ablação por Cateter , Linhagem Celular Tumoral , Citometria de Fluxo , Células-Tronco Hematopoéticas/patologia , Hepatectomia/métodos , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Imuno-Histoquímica , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Invasividade Neoplásica/patologia , Micrometástase de Neoplasia/patologia , Recidiva Local de Neoplasia/metabolismo , Neoplasia Residual/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Tirapazamina , Triazinas/uso terapêutico
5.
EMBO Rep ; 12(9): 931-7, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21760611

RESUMO

The death receptor CD95 promotes apoptosis through well-defined signalling pathways. In colorectal cancer cells, CD95 primarily stimulates migration and invasion through pathways that are incompletely understood. Here, we identify a new CD95-activated tyrosine kinase pathway that is essential for CD95-stimulated tumour cell invasion. We show that CD95 promotes Tyr 783 phosphorylation of phospholipase C-γ1 through the platelet-derived growth factor receptor-ß, resulting in ligand-stimulated phosphatidylinositol (4,5)-bisphosphate (PIP(2)) hydrolysis. PIP(2) hydrolysis liberates the actin-severing protein cofilin from the plasma membrane to initiate cortical actin remodelling. Cofilin activation is required for CD95-stimulated formation of membrane protrusions and increased tumour cell invasion.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Neoplasias Colorretais/metabolismo , Fosfatidilinositóis/metabolismo , Transdução de Sinais , Receptor fas/metabolismo , Actinas/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Extensões da Superfície Celular , Neoplasias Colorretais/patologia , Camundongos , Invasividade Neoplásica , Fosfolipase C gama/metabolismo , Fosforilação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Morte Celular/metabolismo
6.
Gastroenterology ; 141(1): 269-78, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21459094

RESUMO

BACKGROUND & AIMS: Stem cells of normal tissues have resistance mechanisms that allow them to survive genotoxic insults. The stem cell-like cells of tumors are defined by their tumor-initiating capacity and may have retained these resistance mechanisms, making them resistant to chemotherapy. We studied the relationship between resistance to the topoisomerase I inhibitor irinotecan and tumor-initiating potential in human colonosphere cultures and in mice with colorectal xenograft tumors. METHODS: Colonosphere cultures were established from human colorectal tumor specimens obtained from patients who underwent colon or liver resection for primary or metastatic adenocarcinoma. Stem cell and differentiation markers were analyzed by immunoblotting and fluorescence-activated cell sorting. Clone- and tumor-initiating capacities were assessed by single-cell cloning and in immune-deficient mice. Sensitivity to irinotecan was assessed in vitro and in tumor-bearing mice. The relationship between drug resistance and tumor-initiating capacity was tested by fluorescence-activated cell sorting of colonosphere cells, based on expression of ABCB1 and aldehyde dehydrogenase (ALDH) activity. RESULTS: Colonosphere cultures had a high capacity to initiate tumors in mice and were resistant to irinotecan. Inhibition of the drug-efflux pump ABCB1 by PSC-833 allowed irinotecan to eradicate tumor-initiating cells. However, ABCB1 was expressed only by a subpopulation of differentiated tumor cells that did not form clones or tumors. Conversely, tumor-initiating cells were ABCB1-negative and were identified by high ALDH activity. Tumorigenic ALDHhigh/ABCB1negative cells generated nontumorigenic ALDHlow/ABCB1positive daughter cells in vitro and in tumor xenografts. PSC-833 increased the antitumor efficacy of irinotecan in mice. CONCLUSIONS: The resistance of colorectal tumors to irinotecan requires the cooperative action of tumor-initiating ALDHhigh/ABCB1negative cells and their differentiated, drug-expelling, ALDHlow/ABCB1positive daughter cells.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/análogos & derivados , Diferenciação Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Neoplasias Hepáticas/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Inibidores da Topoisomerase I/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Aldeído Desidrogenase/metabolismo , Animais , Antineoplásicos Fitogênicos/metabolismo , Biomarcadores Tumorais/metabolismo , Western Blotting , Camptotecina/metabolismo , Camptotecina/farmacologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ciclosporinas/farmacologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Citometria de Fluxo/métodos , Humanos , Irinotecano , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Esferoides Celulares , Fatores de Tempo , Inibidores da Topoisomerase I/metabolismo , Carga Tumoral/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Cell Mol Med ; 15(10): 2117-29, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21155976

RESUMO

Stromal precursor antigen (STRO)-3 has previously been shown to identify a subset of adult human bone marrow (BM)-derived mesenchymal lineage precursors, which may have cardioprotective potential. We sought to characterize STRO-3-immunoselected and culture-expanded mesenchymal precursor cells (MPCs) with respect to their biology and therapeutic potential in myocardial ischemia. Immunoselection of STRO-3(+) MPCs enriched for fibroblastic colony forming units from unfractionated BM mononuclear cells (MNCs). Compared to mesenchymal stem cells conventionally isolated by plastic adherence, MPCs demonstrated increased proliferative capacity during culture expansion, expressed higher levels of early 'stem cell' markers and various pro-angiogenic and cardioprotective cytokines, and exhibited greater trilineage developmental efficiency. Intramyocardial injection of MPCs into a rat model of myocardial infarction (MI) promoted left ventricular recovery and inhibited left ventricular dilatation. These beneficial effects were associated with cardioprotective and pro-angiogenic effects at the tissue level, despite poor engraftment of cells. Treatment of MI rats with MPC-conditioned medium (CM) preserved left ventricular function and dimensions, reduced myocyte apoptosis and fibrosis, and augmented neovascularization, involving both resident vascular cells and circulating endothelial progenitor cells (EPCs). Profiling of CM revealed various cardioprotective and pro-angiogenic factors, which had biological activity in cultures of myocytes, tissue-resident vascular cells and EPCs. Prospective immunoselection of STRO-3(+) MPCs from BM MNCs conferred advantage in maintaining a population of immature MPCs during ex vivo expansion. Transplantation of culture-expanded MPCs into the post-MI heart resulted in therapeutic benefit, attributable at least in part to paracrine mechanisms of action. Thus, MPCs represent a promising therapy for myocardial ischemia.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Isquemia Miocárdica/terapia , Animais , Antígenos/análise , Medula Óssea/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Ratos Nus , Células Estromais/metabolismo
8.
J Surg Oncol ; 104(2): 198-204, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21506133

RESUMO

BACKGROUND: Ischemia/reperfusion (I/R) injury in the liver is associated with accelerated outgrowth of micrometastases. The aim of the study was to test the role of CD95 signaling in accelerated outgrowth of colorectal liver metastases following I/R. METHODS: Mice underwent vascular clamping 5 days after induction of colorectal liver metastases. Invasion and outgrowth of micrometastases following I/R were analyzed by post-mortem confocal microscopy (36 hr post-I/R) and by morphometric assessment of tumor load (5 days post-I/R), respectively. Tumor cell CD95 was suppressed by lentiviral RNA interference. The contribution of host CD95L was assessed by using gld-mice lacking functional CD95L. RESULTS: CD95-knockdown in tumor cells strongly reduced perinecrotic invasion (tumor diameter from ∼830 to ∼470 µm) and largely prevented outgrowth acceleration of perinecrotic liver metastases following I/R (from ∼8- to ∼4.5-fold). In gld-mice, the relative hepatic area with necrosis was markedly reduced. Perinecrotic tumor cell clusters still displayed an invasive phenotype (tumor diameter of ∼980 µm). However, I/R-induced acceleration of tumor outgrowth was reduced in gld-mice from ∼8- to ∼5-fold. CONCLUSIONS: I/R induces invasion and accelerated outgrowth of preestablished metastases in a CD95-dependent manner. Activation of the CD95 system following I/R not only contributes to liver injury, but may also promote aggressive tumor recurrence.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Traumatismo por Reperfusão/metabolismo , Receptor fas/metabolismo , Animais , Neoplasias Colorretais/patologia , Fígado/irrigação sanguínea , Fígado/patologia , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais
9.
J Surg Oncol ; 101(7): 551-6, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20461760

RESUMO

BACKGROUND: Recently, we have shown in a murine model that radiofrequency ablation (RFA) induces accelerated outgrowth of colorectal micrometastases in the transition zone (TZ) surrounding the ablated lesion. Conversely, RFA also induces an anti-tumor T-cell response that may limit tumor growth at distant sites. Here we have evaluated whether an altered density of inflammatory cells could be observed in the perinecrotic (TZ) metastases compared to hepatic metastases in the distant reference zone (RZ). METHODS: RFA-treated tumor-bearing mice (n = 10) were sacrificed. The inflammatory cell density (neutrophils, macrophages, CD4(+) T-cells, and CD8(+) T-cells) of tumors in the TZ (TZ tumors) was compared to that in tumors in the RZ (RZ tumors). Sham-operated, tumor-bearing mice (n = 10) were analyzed simultaneously as controls (sham-treated tumors). RESULTS: In RFA-treated, tumor-bearing mice RZ tumors contained a significantly higher density of neutrophils and CD4(+) T-cells, but not macrophages and CD8(+) T-cells compared to sham-treated tumors. Notably, TZ tumors had a significantly lower density of neutrophils, CD4(+) T-cells, and CD8(+) T-cells, but not macrophages, when compared to RZ tumors. CONCLUSIONS: The accelerated perinecrotic tumor outgrowth following RFA is associated with a reduced density of neutrophils and T-cells compared to distant hepatic metastases. This may have implications for local tumor recurrence following RFA.


Assuntos
Ablação por Cateter/efeitos adversos , Neoplasias Colorretais/patologia , Inflamação/etiologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Recidiva Local de Neoplasia/patologia , Animais , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/metabolismo , Inflamação/patologia , Neoplasias Hepáticas/secundário , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/metabolismo
10.
Surg Oncol ; 26(4): 535-543, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29113675

RESUMO

Metastatic colorectal cancer (mCRC) is a devastating disease causing 700.000 deaths annually worldwide. Metastases most frequently develop in the liver. Partial hepatectomy has dramatically improved clinical outcome and is the only curative treatment option for eligible patients with mCRC. Pre-clinical studies have shown that surgical procedures can have tumor-promoting local 'side-effects' such as hypoxia and inflammation, thereby altering the behaviour of residual tumor cells. In addition, systemically released factors following (colon or liver) surgery can act as a wakeup-call for dormant tumor cells in distant organs and/or help establish a pre-metastatic niche. Tumor handling during resection may also increase the number of circulating tumor cells. Despite the overwhelming amount of pre-clinical data demonstrating the pro-tumorigenic side effects of surgery, clinical evidence is scarce. Indications for hepatic surgery are rapidly increasing due to a rise in the incidence of mCRC and a trend towards more aggressive surgical treatment. Therefore, it is increasingly important to understand the principles of surgery-induced tumor growth, in order to devise perioperative or adjuvant strategies to further enhance long-term tumor control. In the current study we review the evidence for surgery-stimulated tumor growth and suggest strategies to assess the clinical relevance of such findings.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Hepatectomia/efeitos adversos , Neoplasias Hepáticas/secundário , Humanos , Neoplasias Hepáticas/etiologia , Prognóstico
11.
Oncotarget ; 8(49): 86296-86311, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29156796

RESUMO

Surgical removal of colorectal cancer (CRC) liver metastases generates areas of tissue hypoxia. Hypoxia imposes a stem-like phenotype on residual tumor cells and promotes tumor recurrence. Moreover, in primary CRC, gene expression signatures reflecting hypoxia and a stem-like phenotype are highly expressed in the aggressive Consensus Molecular Subtype 4 (CMS4). Therapeutic strategies eliminating hypoxic stem-like cells may limit recurrence following resection of primary tumors or metastases. Here we show that expression of DNA repair genes is strongly suppressed in CMS4 and inversely correlated with hypoxia-inducible factor-1 alpha (HIF1α) and HIF-2α co-expression signatures. Tumors with high expression of HIF signatures and low expression of repair proteins showed the worst survival. In human tumors, expression of the repair proteins RAD51, KU70 and RIF1 was strongly suppressed in hypoxic peri-necrotic tumor areas. Experimentally induced hypoxia in patient derived colonospheres in vitro or in vivo (through vascular clamping) was sufficient to downregulate repair protein expression and caused DNA damage. Hypoxia-induced DNA damage was prevented by expressing the hydroperoxide-scavenging enzyme glutathione peroxidase-2 (GPx2), indicating that reactive oxygen species mediate hypoxia-induced DNA damage. Finally, the hypoxia-activated prodrug Tirapazamine greatly augmented DNA damage and reduced the fraction of stem-like (Aldefluorbright) tumor cells in vitro, and in vivo following vascular clamping. We conclude that decreased expression of DNA repair proteins and increased DNA damage in hypoxic tumor areas may be therapeutically exploited with hypoxia-activated prodrugs, and that such drugs reduce the fraction of Aldefluorbright (stem-like) tumor cells.

12.
Oncotarget ; 7(34): 54662-54675, 2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27323406

RESUMO

The long term prognosis of liver cancer patients remains unsatisfactory because of cancer recurrence after surgical interventions, particularly in patients with viral infections. Since hepatitis B and C viral proteins lead to inactivation of the tumor suppressors p53 and Retinoblastoma (Rb), we hypothesize that surgery in the context of p53/Rb inactivation initiate de novo tumorigenesis.We, therefore, generated transgenic mice with hepatocyte and cholangiocyte/liver progenitor cell (LPC)-specific deletion of p53 and Rb, by interbreeding conditional p53/Rb knockout mice with either Albumin-cre or Cytokeratin-19-cre transgenic mice.We show that liver cancer develops at the necrotic injury site after surgical resection or radiofrequency ablation in p53/Rb deficient livers. Cancer initiation occurs as a result of specific migration, expansion and transformation of cytokeratin-19+-liver (CK-19+) cells. At the injury site migrating CK-19+ cells formed small bile ducts and adjacent cells strongly expressed the transforming growth factor ß (TGFß). Isolated cytokeratin-19+ cells deficient for p53/Rb were resistant against hypoxia and TGFß-mediated growth inhibition. CK-19+ specific deletion of p53/Rb verified that carcinomas at the injury site originates from cholangiocytes or liver progenitor cells.These findings suggest that human liver patients with hepatitis B and C viral infection or with mutations for p53 and Rb are at high risk to develop tumors at the surgical intervention site.


Assuntos
Transformação Celular Neoplásica/genética , Queratina-19/genética , Neoplasias Hepáticas/genética , Fígado/metabolismo , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética , Animais , Ablação por Cateter/efeitos adversos , Ablação por Cateter/métodos , Proliferação de Células/genética , Transformação Celular Neoplásica/metabolismo , Hepatectomia/efeitos adversos , Hepatectomia/métodos , Hepatócitos/metabolismo , Humanos , Queratina-19/metabolismo , Fígado/patologia , Fígado/cirurgia , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína do Retinoblastoma/deficiência , Técnicas de Cultura de Tecidos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteína Supressora de Tumor p53/deficiência
13.
Cancer Res ; 74(22): 6717-30, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25261240

RESUMO

Colorectal tumorigenesis is accompanied by the generation of oxidative stress, but how this controls tumor development is poorly understood. Here, we studied how the H2O2-reducing enzyme glutathione peroxidase 2 (GPx2) regulates H2O2 stress and differentiation in patient-derived "colonosphere" cultures. GPx2 silencing caused accumulation of radical oxygen species, sensitization to H2O2-induced apoptosis, and strongly reduced clone- and metastasis-forming capacity. Neutralization of radical oxygen species restored clonogenic capacity. Surprisingly, GPx2-suppressed cells also lacked differentiation potential and formed slow-growing undifferentiated tumors. GPx2 overexpression stimulated multilineage differentiation, proliferation, and tumor growth without reducing the tumor-initiating capacity. Finally, GPx2 expression was inversely correlated with H2O2-stress signatures in human colon tumor cohorts, but positively correlated with differentiation and proliferation. Moreover, high GPx2 expression was associated with early tumor recurrence, particularly in the recently identified aggressive subtype of human colon cancer. We conclude that H2O2 neutralization by GPx2 is essential for maintaining clonogenic and metastatic capacity, but also for the generation of differentiated proliferating tumor mass. The results reveal an unexpected redox-controlled link between tumor mass formation and metastatic capacity.


Assuntos
Neoplasias Colorretais/patologia , Glutationa Peroxidase/fisiologia , Peróxido de Hidrogênio/metabolismo , Animais , Diferenciação Celular , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Camundongos , Camundongos SCID , Metástase Neoplásica , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico , Tiorredoxina Redutase 1/fisiologia
14.
Nat Commun ; 5: 5275, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25387467

RESUMO

Host responses to systemic anti-cancer treatment play important roles in the development of anti-cancer drug resistance. Here we show that F4/80(+)/CD11b(low) splenocytes mediate the resistance to DNA-damaging chemotherapeutics induced by two platinum-induced fatty acids (PIFAs), 12-S-keto-5,8,10-heptadecatrienoic acid and 4,7,10,13-hexadecatetraenoic acid (16:4(n-3)) in xenograft mouse models. Splenectomy or depletion of splenic macrophages by liposomal clodronate protects against PIFA-induced chemoresistance. In addition, we find that 12-S-HHT, but not 16:4(n-3), functions via leukotriene B4 receptor 2 (BLT2). Genetic loss or chemical inhibition of BLT2 prevents 12-S-HHT-mediated resistance. Mass spectrometry analysis of conditioned medium derived from PIFA-stimulated splenic macrophages identifies several lysophosphatidylcholines as the resistance-inducing molecules. When comparing cisplatin and PIFA-treated tumours with cisplatin alone treated tumours we found overall less γH2AX, a measure for DNA damage. Taken together, we have identified an intricate network of lysophospholipid signalling by splenic macrophages that induces systemic chemoresistance in vivo via an altered DNA damage response.


Assuntos
Dano ao DNA , Resistencia a Medicamentos Antineoplásicos/fisiologia , Lisofosfolipídeos/fisiologia , Macrófagos/metabolismo , Animais , Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Dano ao DNA/fisiologia , Ácidos Graxos Insaturados/metabolismo , Ácidos Graxos Insaturados/fisiologia , Lisofosfolipídeos/metabolismo , Macrófagos/fisiologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/tratamento farmacológico , Receptores do Leucotrieno B4/fisiologia , Baço/citologia , Esplenectomia
15.
Neoplasia ; 15(2): 204-17, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23441134

RESUMO

In epithelial tumors, the platelet-derived growth factor receptor B (PDGFRB) is mainly expressed by stromal cells of mesenchymal origin. Tumor cells may also acquire PDGFRB expression following epithelial-to-mesenchymal transition (EMT), which occurs during metastasis formation. Little is known about PDGFRB signaling in colorectal tumor cells. We studied the relationship between PDGFRB expression, EMT, and metastasis in human colorectal cancer (CRC) cohorts by analysis of gene expression profiles. PDGFRB expression in primary CRC was correlated with short disease-free and overall survival. PDGFRB was co-expressed with genes involved in platelet activation, transforming growth factor beta (TGFB) signaling, and EMT in three CRC cohorts. PDGFRB was expressed in mesenchymal-like tumor cell lines in vitro and stimulated invasion and liver metastasis formation in mice. Platelets, a major source of PDGF, preferentially bound to tumor cells in a non-activated state. Platelet activation caused robust PDGFRB tyrosine phosphorylation on tumor cells in vitro and in liver sinusoids in vivo. Platelets also release TGFB, which is a potent inducer of EMT. Inhibition of TGFB signaling in tumor cells caused partial reversion of the mesenchymal phenotype and strongly reduced PDGFRB expression and PDGF-stimulated tumor cell invasion. These results suggest that PDGFRB may contribute to the aggressive phenotype of colorectal tumors with mesenchymal properties, most likely downstream of platelet activation and TGFB signaling.


Assuntos
Neoplasias Colorretais/genética , Transição Epitelial-Mesenquimal/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Transformador beta/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/secundário , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Ativação Plaquetária/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Transcriptoma/genética , Fator de Crescimento Transformador beta/metabolismo
16.
J Proteomics ; 91: 84-96, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-23835434

RESUMO

Drug-resistant cancer stem cells (CSCs) have been implicated in tumor recurrence following chemotherapy. However, the contribution of CSCs to drug-resistance in colorectal cancer is unclear and CSC-intrinsic drug-resistance mechanisms are ill-defined. Here, we address these issues by proteomic analysis of the secretomes of CSCs and isogenic differentiated tumor cells (DTCs) isolated from three distinct metastasized colon tumors. Mass spectrometry-based proteomics identified 1254 unique proteins in the conditioned media of the paired CSC and DTC cultures. Ingenuity Pathway Analysis revealed that proteins governing 'Cell Death' were most significantly enriched in the CSC secretome. The vast majority of these (37/43) promote cell survival. The CSC secretome is also characterized by a pro-survival Nrf2 antioxidant signature. Interestingly, proteome-maintenance networks are highly enriched in the CSC secretome. CSCs also secrete high levels of drug-metabolizing enzymes, including aldehyde dehydrogenase 1 (ALDH1A1) and bleomycin hydrolase (BLMH). We show that these enzymes cause extracellular detoxification of maphosphamide and bleomycin respectively. We conclude that colorectal CSCs are characterized by extensive survival and anti-oxidant networks, which are likely to contribute to CSC-intrinsic drug-resistance. In addition, CSCs may modulate drug responses in nearby tumor cells by detoxifying chemotherapeutic drugs in the extracellular space. BIOLOGICAL SIGNIFICANCE: Cancer stem cells are thought to play an important role in mediating drug resistance and tumor recurrence following chemotherapy. Therefore, it is important to identify the factors that are secreted by them. Our results provide novel insights into the pathways that govern the intrinsic resistance of CSCs to chemotherapy and, furthermore, demonstrate that they can also inactivate chemotherapeutic drugs in the extracellular space. A better understanding of the pathways that govern drug resistance in CSCs may help in developing effective CSC-targeting drugs.


Assuntos
Neoplasias do Colo/metabolismo , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Proteoma/metabolismo , Antineoplásicos/farmacologia , Antioxidantes/química , Antioxidantes/metabolismo , Bleomicina/farmacologia , Linhagem Celular Tumoral , Separação Celular , Sobrevivência Celular , Biologia Computacional , Meios de Cultivo Condicionados/química , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacologia , Humanos , Células-Tronco Neoplásicas/metabolismo , Estresse Oxidativo , Dobramento de Proteína , Proteômica , Recidiva
17.
Curr Colorectal Cancer Rep ; 8(2): 130-137, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22611343

RESUMO

In patients with unresectable colorectal liver metastases (CRLM), radiofrequency ablation (RFA) might be a good alternative, whenever possible. In contrast to systemic therapy, the aim of RFA is to achieve complete local tumor control in an attempt to provide long-term survival. In this article we discuss the available evidence regarding the treatment of patients with unresectable CRLM, focusing on RFA in conjunction with modern systemic therapies. We observed that the available evidence in the existing literature is limited, and often consists of level 2 and 3 evidence, thereby hampering any firm conclusions. Nonetheless, RFA seems superior to chemotherapy alone in patients with liver-only disease amenable for RFA. However, the combination of RFA and chemotherapy has been demonstrated to be feasible and safe, lending support to the concept of RFA followed by chemotherapy, in order to reduce local recurrence rates and prolong survival.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA