Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Proc Natl Acad Sci U S A ; 114(41): E8685-E8694, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973887

ABSTRACT

The molecular underpinnings of invasion, a hallmark of cancer, have been defined in terms of individual mediators but crucial interactions between these mediators remain undefined. In xenograft models and patient specimens, we identified a c-Met/ß1 integrin complex that formed during significant invasive oncologic processes: breast cancer metastases and glioblastoma invasive resistance to antiangiogenic VEGF neutralizing antibody, bevacizumab. Inducing c-Met/ß1 complex formation through an engineered inducible heterodimerization system promoted features crucial to overcoming stressors during metastases or antiangiogenic therapy: migration in the primary site, survival under hypoxia, and extravasation out of circulation. c-Met/ß1 complex formation was up-regulated by hypoxia, while VEGF binding VEGFR2 sequestered c-Met and ß1 integrin, preventing their binding. Complex formation promoted ligand-independent receptor activation, with integrin-linked kinase phosphorylating c-Met and crystallography revealing the c-Met/ß1 complex to maintain the high-affinity ß1 integrin conformation. Site-directed mutagenesis verified the necessity for c-Met/ß1 binding of amino acids predicted by crystallography to mediate their extracellular interaction. Far-Western blotting and sequential immunoprecipitation revealed that c-Met displaced α5 integrin from ß1 integrin, creating a complex with much greater affinity for fibronectin (FN) than α5ß1. Thus, tumor cells adapt to microenvironmental stressors induced by metastases or bevacizumab by coopting receptors, which normally promote both cell migration modes: chemotaxis, movement toward concentrations of environmental chemoattractants, and haptotaxis, movement controlled by the relative strengths of peripheral adhesions. Tumor cells then redirect these receptors away from their conventional binding partners, forming a powerful structural c-Met/ß1 complex whose ligand-independent cross-activation and robust affinity for FN drive invasive oncologic processes.


Subject(s)
Breast Neoplasms/secondary , Drug Resistance, Neoplasm , Glioblastoma/secondary , Integrin beta1/metabolism , Proto-Oncogene Proteins c-met/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Bevacizumab/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Female , Fibronectins/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Integrin beta1/genetics , Mice , Neoplasm Invasiveness , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
JCI Insight ; 2(2): e88815, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138554

ABSTRACT

Clinical trials revealed limited response duration of glioblastomas to VEGF-neutralizing antibody bevacizumab. Thriving in the devascularized microenvironment occurring after antiangiogenic therapy requires tumor cell adaptation to decreased glucose, with 50% less glucose identified in bevacizumab-treated xenografts. Compared with bevacizumab-responsive xenograft cells, resistant cells exhibited increased glucose uptake, glycolysis, 13C NMR pyruvate to lactate conversion, and survival in low glucose. Glucose transporter 3 (GLUT3) was upregulated in bevacizumab-resistant versus sensitive xenografts and patient specimens in a HIF-1α-dependent manner. Resistant versus sensitive cell mitochondria in oxidative phosphorylation-selective conditions produced less ATP. Despite unchanged mitochondrial numbers, normoxic resistant cells had lower mitochondrial membrane potential than sensitive cells, confirming poorer mitochondrial health, but avoided the mitochondrial dysfunction of hypoxic sensitive cells. Thin-layer chromatography revealed increased triglycerides in bevacizumab-resistant versus sensitive xenografts, a change driven by mitochondrial stress. A glycogen synthase kinase-3ß inhibitor suppressing GLUT3 transcription caused greater cell death in bevacizumab-resistant than -responsive cells. Overexpressing GLUT3 in tumor cells recapitulated bevacizumab-resistant cell features: survival and proliferation in low glucose, increased glycolysis, impaired oxidative phosphorylation, and rapid in vivo proliferation only slowed by bevacizumab to that of untreated bevacizumab-responsive tumors. Targeting GLUT3 or the increased glycolysis reliance in resistant tumors could unlock the potential of antiangiogenic treatments.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glucose Transporter Type 3/genetics , Glycolysis , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab/pharmacology , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm/drug effects , Glioblastoma/blood supply , Glioblastoma/genetics , Glioblastoma/metabolism , Glucose/metabolism , Glucose Transporter Type 3/drug effects , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Phosphorylation , Pyruvic Acid/metabolism , Up-Regulation
3.
Am J Kidney Dis ; 69(2): 192-199, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27522513

ABSTRACT

BACKGROUND: Cardiac surgery-related acute kidney injury (AKI) is a common postoperative complication that greatly increases morbidity and mortality. There are currently no effective interventions to prevent AKI associated with cardiac surgery. Experimental data have shown that administration of the mineralocorticoid receptor blocker spironolactone prevents renal injury induced by ischemia-reperfusion in rats. The objective of this study was to test whether short-term perioperative administration of oral spironolactone could reduce the incidence of AKI in cardiac surgical patients. STUDY DESIGN: Randomized, double-blinded, placebo-controlled trial. SETTING & PARTICIPANTS: Data were collected from April 2014 through July 2015 at the National Heart Institute in Mexico. 233 patients were included; 115 and 118 received spironolactone or placebo, respectively. INTERVENTION: Spironolactone or placebo once at a dose of 100mg 12 to 24 hours before surgery and subsequently 3 further doses of 25mg in postoperative days 0, 1, and 2 were administered. OUTCOMES: Patients were followed up for 7 days or until discharge from the intensive care unit (ICU). The primary end point was AKI incidence defined by KDIGO criteria. Secondary end points included requirement of renal replacement therapy, ICU length of stay, and ICU mortality. Data were analyzed according to the intention-to-treat principle. RESULTS: Mean age was 53.2±15 years, mean serum creatinine level was 0.9±0.2mg/dL, median Thakar score for estimation of AKI risk was 2 (IQR, 1-3), and 25% had diabetes. The incidence of AKI was higher for the spironolactone group (43% vs 29%; P=0.02). No significant differences were found for secondary end points. LIMITATIONS: Single center, AKI was mostly driven by AKI stage 1, planned sample size was not achieved, and there was no renin-angiotensin-aldosterone system washout period. CONCLUSIONS: Our trial demonstrated that spironolactone was not protective for AKI associated with cardiac surgery and there may be a trend toward risk.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Cardiac Surgical Procedures/adverse effects , Mineralocorticoid Receptor Antagonists/therapeutic use , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Spironolactone/therapeutic use , Double-Blind Method , Female , Humans , Intensive Care Units , Male , Middle Aged
4.
Clin Cancer Res ; 19(7): 1773-83, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23307858

ABSTRACT

PURPOSE: To identify mediators of glioblastoma antiangiogenic therapy resistance and target these mediators in xenografts. EXPERIMENTAL DESIGN: We conducted microarray analysis comparing bevacizumab-resistant glioblastomas (BRG) with pretreatment tumors from the same patients. We established novel xenograft models of antiangiogenic therapy resistance to target candidate resistance mediator(s). RESULTS: BRG microarray analysis revealed upregulation versus pretreatment of receptor tyrosine kinase c-Met, which underwent further investigation because of its prior biologic plausibility as a bevacizumab resistance mediator. BRGs exhibited increased hypoxia versus pretreatment in a manner correlating with their c-Met upregulation, increased c-Met phosphorylation, and increased phosphorylation of c-Met-activated focal adhesion kinase and STAT3. We developed 2 novel xenograft models of antiangiogenic therapy resistance. In the first model, serial bevacizumab treatment of an initially responsive xenograft generated a xenograft with acquired bevacizumab resistance, which exhibited upregulated c-Met expression versus pretreatment. In the second model, a BRG-derived xenograft maintained refractoriness to the MRI tumor vasculature alterations and survival-promoting effects of bevacizumab. Growth of this BRG-derived xenograft was inhibited by a c-Met inhibitor. Transducing these xenograft cells with c-Met short hairpin RNA inhibited their invasion and survival in hypoxia, disrupted their mesenchymal morphology, and converted them from bevacizumab-resistant to bevacizumab-responsive. Engineering bevacizumab-responsive cells to express constitutively active c-Met caused these cells to form bevacizumab-resistant xenografts. CONCLUSION: These findings support the role of c-Met in survival in hypoxia and invasion, features associated with antiangiogenic therapy resistance, and growth and therapeutic resistance of xenografts resistant to antiangiogenic therapy. Therapeutically targeting c-Met could prevent or overcome antiangiogenic therapy resistance.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Transcriptome , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cluster Analysis , Drug Resistance, Neoplasm/genetics , Enzyme Activation/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Mice , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA Interference , Xenograft Model Antitumor Assays
5.
Rev Invest Clin ; 65(6): 467-75, 2013.
Article in English | MEDLINE | ID: mdl-24687353

ABSTRACT

INTRODUCTION: Acute kidney injury (AKI) associated with cardiac surgery is a common postoperative complication that increases the morbidity and mortality substantially. However, there is limited information of AKI after cardiac surgery in our institution. MATERIAL AND METHODS: We conducted a prospective, observational, and longitudinal analysis of adult patients that underwent to cardiac surgery requiring cardiopulmonary bypass and aortic cross clamp. Patients with preoperative chronic renal insufficiency that were on dialysis, with AKI detected up to 24 h before the procedure, or that received contrast agents 72 h before surgery were excluded. AKI was defined by the AKIN classification. Patients were followed up to 7 days after surgery or before if discharged from the intensive care unit. We analyzed age, sex, body mass index (BMI), co-morbilities, previous cardiac surgery, left ventricular ejection fraction, New York Heart Association class, type of procedure, cardiopulmonary bypass time, cross clamp time and bleeding. RESULTS: Our analysis included 164 patients submitted to cardiac surgery. In the follow up, 84% did not have AKI, 11% had AKIN 1 and 2 accompanied by increase in serum creatinine and 6% had AKIN 3. Patients with AKI were older, had a higher preoperative creatinine, plasma glucose level, and a lower left ventricular ejection fraction. All together patients with AKIN had a longer hospital stay and a higher mortality (p < 0.001). The preoperative use of insulin was associated with the development of AKI, and there was a higher number of patients with a New York Heart Association class III and IV for heart failure in the more sever forms of AKI (p = 0.01). The logistic regression analysis revealed that patients with a high preoperative blood urea nitrogen (> 20 mg/dL) creatinine level (> 1 mg/dL), uric acid (> 7 mg/dL) and lower albumin (< 4 g/dL) or lower intraoperative hemoglobin (< 8 g/dL) had a higher risk for postoperative AKI. CONCLUSIONS: The prevalence of AKI in our Institute is of 17%. Patients with AKIN 2 and 3 had a higher mortality and a longer stay in the intensive care unit. The major risk factors for AKI development were identified.


Subject(s)
Acute Kidney Injury/epidemiology , Cardiac Surgical Procedures , Postoperative Complications/epidemiology , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Acute Kidney Injury/therapy , Adult , Age Factors , Aged , Blood Glucose/analysis , Body Mass Index , Cardiopulmonary Bypass , Creatinine/blood , Diabetes Complications/drug therapy , Diabetes Complications/epidemiology , Elective Surgical Procedures , Female , Follow-Up Studies , Humans , Intensive Care Units/statistics & numerical data , Length of Stay/statistics & numerical data , Male , Mexico/epidemiology , Middle Aged , Postoperative Complications/blood , Postoperative Complications/etiology , Postoperative Complications/therapy , Prospective Studies , Renal Replacement Therapy/statistics & numerical data , Risk Factors , Serum Albumin/analysis , Sex Factors , Stroke Volume
6.
Autophagy ; 8(6): 979-81, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22714142

ABSTRACT

While anti-angiogenic therapy was initially greeted enthusiastically by the cancer community, initial successes with this therapeutic modality were tempered by the failure of angiogenesis inhibitors to produce sustained clinical responses in most patients, with resistance to the inhibitors frequently developing. We recently reported that hypoxia increases after the devascularization caused by anti-angiogenic therapy, consistent with the goals of these therapies, but that some tumor cells become resistant and survive the hypoxic insult elicited by anti-angiogenic therapy through autophagy by activating both AMPK and HIF1A pathways. These findings suggest that modulating the autophagy pathway may someday allow anti-angiogenic therapy to fulfill its therapeutic potential. However, further work will clearly be needed to develop more potent and specific autophagy inhibitors and to better understand the regulators of autophagy in malignant cells.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Autophagy , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Autophagy/drug effects , Cell Hypoxia/drug effects , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Biological , Neoplasms/blood supply , Neoplastic Stem Cells/pathology , Tumor Microenvironment/drug effects
7.
Mol Cell ; 37(5): 620-32, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20227367

ABSTRACT

To sustain tumor growth, cancer cells must be able to adapt to fluctuations in energy availability. We have identified a single microRNA that controls glioma cell proliferation, migration, and responsiveness to glucose deprivation. Abundant glucose allows relatively high miR-451 expression, promoting cell growth. In low glucose, miR-451 levels decrease, slowing proliferation but enhancing migration and survival. This allows cells to survive metabolic stress and seek out favorable growth conditions. In glioblastoma patients, elevated miR-451 is associated with shorter survival. The effects of miR-451 are mediated by LKB1, which it represses through targeting its binding partner, CAB39 (MO25 alpha). Overexpression of miR-451 sensitized cells to glucose deprivation, suggesting that its downregulation is necessary for robust activation of LKB1 in response to metabolic stress. Thus, miR-451 is a regulator of the LKB1/AMPK pathway, and this may represent a fundamental mechanism that contributes to cellular adaptation in response to altered energy availability.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain Neoplasms/enzymology , Glioblastoma/enzymology , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stress, Physiological , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/genetics , Adaptation, Physiological , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , COS Cells , Calcium-Binding Proteins/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Chlorocebus aethiops , Enzyme Activation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Glucose/deficiency , HeLa Cells , Humans , Prognosis , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Stress, Physiological/genetics , Time Factors , Transfection
8.
Blood ; 108(4): 1334-8, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16670263

ABSTRACT

Drug development in human chronic lymphocytic leukemia (CLL) has been limited by lack of a suitable animal model to adequately assess pharmacologic properties relevant to clinical application. A recently described TCL-1 transgenic mouse develops a chronic B-cell CD5(+) leukemia that might be useful for such studies. Following confirmation of the natural history of this leukemia in the transgenic mice, we demonstrated that the transformed murine lymphocytes express relevant therapeutic targets (Bcl-2, Mcl-1, AKT, PDK1, and DNMT1), wild-type p53 status, and in vitro sensitivity to therapeutic agents relevant to the treatment of human CLL. We then demonstrated the in vivo clinical activity of low-dose fludarabine in transgenic TCL-1 mice with active leukemia. These studies demonstrated both early reduction in blood-lymphocyte count and spleen size and prolongation of survival (P = .046) compared with control mice. Similar to human CLL, an emergence of resistance was noted with fludarabine treatment in vivo. Overall, these studies suggest that the TCL-1 transgenic leukemia mouse model has similar clinical and therapeutic response properties to human CLL and may therefore serve as a useful in vivo tool to screen new drugs for subsequent development in CLL.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Neoplasm Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Vidarabine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , CD5 Antigens/metabolism , Cell Transformation, Neoplastic/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Mice , Mice, Transgenic , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Cells, Cultured , Vidarabine/pharmacology , Vidarabine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...