Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Int J Mol Sci ; 24(8)2023 Apr 11.
Article in English | MEDLINE | ID: mdl-37108234

ABSTRACT

Therapeutic oligonucleotides are powerful tools for the inhibition of potential targets involved in cancer. We describe the effect of two Polypurine Reverse Hoogsteen (PPRH) hairpins directed against the ERBB2 gene, which is overexpressed in positive HER-2 breast tumors. The inhibition of their target was analyzed by cell viability and at the mRNA and protein levels. The combination of these specific PPRHs with trastuzumab was also explored in breast cancer cell lines, both in vitro and in vivo. PPRHs designed against two intronic sequences of the ERBB2 gene decreased the viability of SKBR-3 and MDA-MB-453 breast cancer cells. The decrease in cell viability was associated with a reduction in ERBB2 mRNA and protein levels. In combination with trastuzumab, PPRHs showed a synergic effect in vitro and reduced tumor growth in vivo. These results represent the preclinical proof of concept of PPRHs as a therapeutic tool for breast cancer.


Subject(s)
Breast Neoplasms , Genes, erbB-2 , Humans , Female , Trastuzumab/pharmacology , Trastuzumab/genetics , Oncogenes , MCF-7 Cells , RNA, Messenger/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Receptor, ErbB-2/genetics
2.
Int J Cancer ; 147(4): 1163-1179, 2020 08 15.
Article in English | MEDLINE | ID: mdl-31943158

ABSTRACT

Around 40% of newly diagnosed lung cancer patients are Stage IV, where the improvement of survival and reduction of disease-related adverse events is the main goal for oncologists. In this scenario, we present preclinical evidence supporting the use of ABTL0812 in combination with chemotherapy for treating advanced and metastatic Nonsmall cell lung adenocarcinomas (NSCLC) and squamous carcinomas. ABTL0812 is a new chemical entity, currently in Phase 1b/2a clinical trial for advanced squamous NSCLC in combination with paclitaxel and carboplatin (P/C), after successfully completing the first-in-human trial where it showed an excellent safety profile and signs of efficacy. We show here that ABTL0812 inhibits Akt/mTOR axis by inducing the overexpression of TRIB3 and activating autophagy in lung squamous carcinoma cell lines. Furthermore, treatment with ABTL0812 also induces AMPK activation and ROS accumulation. Moreover, combination of ABTL0812 with chemotherapy markedly increases the therapeutic effect of chemotherapy without increasing toxicity. We further show that combination of ABTL0812 and chemotherapy induces nonapoptotic cell death mediated by TRIB3 activation and autophagy induction. We also present preliminary clinical data indicating that TRIB3 could serve as a potential novel pharmacodynamic biomarker to monitor ABTL0812 activity administered alone or in combination with chemotherapy in squamous NSCLC patients. The safety profile of ABTL0812 and its good synergy with chemotherapy potentiate the therapeutic potential of current lines of treatment based on chemotherapy regimens, arising as a promising option for improving these patients therapeutic expectancy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Lung Neoplasms/drug therapy , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
3.
Breast Cancer Res ; 20(1): 65, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29973218

ABSTRACT

BACKGROUND: The microenvironment and stress factors like glucocorticoids have a strong influence on breast cancer progression but their role in the first stages of breast cancer and, particularly, in myoepithelial cell regulation remains unclear. Consequently, we investigated the role of glucocorticoids in ductal carcinoma in situ (DCIS) in breast cancer, focusing specially on myoepithelial cells. METHODS: To clarify the role of glucocorticoids at breast cancer onset, we evaluated the effects of cortisol and corticosterone on epithelial and myoepithelial cells using 2D and 3D in vitro and in vivo approaches and human samples. RESULTS: Glucocorticoids induce a reduction in laminin levels and favour the disruption of the basement membrane by promotion of myoepithelial cell apoptosis in vitro. In an in vivo stress murine model, increased corticosterone levels fostered the transition from DCIS to invasive ductal carcinoma (IDC) via myoepithelial cell apoptosis and disappearance of the basement membrane. RU486 is able to partially block the effects of cortisol in vitro and in vivo. We found that myoepithelial cell apoptosis is more frequent in patients with DCIS+IDC than in patients with DCIS. CONCLUSIONS: Our findings show that physiological stress, through increased glucocorticoid blood levels, promotes the transition from DCIS to IDC, particularly by inducing myoepithelial cell apoptosis. Since this would be a prerequisite for invasive features in patients with DCIS breast cancer, its clinical management could help to prevent breast cancer progression to IDC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Ductal, Breast/blood , Carcinoma, Intraductal, Noninfiltrating/blood , Glucocorticoids/blood , Animals , Apoptosis/genetics , Biomarkers, Tumor/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Disease Progression , Female , Heterografts , Humans , Laminin/genetics , Mice , Myoepithelioma/blood , Myoepithelioma/genetics , Myoepithelioma/pathology , Tumor Microenvironment/genetics
4.
Muscle Nerve ; 49(2): 233-48, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23649607

ABSTRACT

INTRODUCTION: Cachexia is a wasting condition that manifests in several types of cancer. The main characteristic of this condition is a profound loss of muscle mass. METHODS: By using a microarray system, expression of several hundred genes was screened in skeletal muscle of rats bearing a cachexia-inducing tumor, the AH-130 Yoshida ascites hepatoma. This model induced a strong decrease in muscle mass in the tumor-bearing animals, as compared with their healthy counterparts. RESULTS: The results show important differences in gene expression in EDL skeletal muscle between tumor-bearing animals with cachexia and control animals. CONCLUSIONS: The differences observed pertain to genes related to intracellular calcium homeostasis and genes involved in the control of mitochondrial oxidative phosphorylation and protein turnover, both at the level of protein synthesis and proteolysis. Assessment of these differences may be a useful tool for the design of novel therapeutic strategies to fight this devastating syndrome.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Excitation Contraction Coupling/physiology , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/physiopathology , Muscle, Skeletal/physiopathology , Animals , Cachexia/etiology , Cachexia/genetics , Cachexia/physiopathology , Calcium/metabolism , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/genetics , Disease Models, Animal , Energy Metabolism/physiology , Excitation Contraction Coupling/genetics , Homeostasis/physiology , Liver Neoplasms/complications , Liver Neoplasms/genetics , Male , Rats , Rats, Wistar
5.
Clin Transl Oncol ; 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898351

ABSTRACT

PURPOSE: To analyze the predictive capacity for local disease control of the transcriptional expression of neogenin-1 (NEO1) gene in patients with head and neck squamous cell carcinoma (HNSCC). METHODS/PATIENTS: A retrospective study was performed on tumor biopsies from 107 patients with HNSCC treated surgically. The transcriptional expression of NEO1 was determined by RT-PCR. NEO1 transcriptional expression value was categorized according to local disease control by recursive partitioning analysis. RESULTS: Lower NEO1 transcriptional expression was associated with worse local control after surgical treatment. Patients with lower NEO1 expression (n = 25, 23.4%) had a 5-year local recurrence-free survival of 61.8% (95% CI: 42.1-81.5%), while patients with higher NEO1 expression (n = 82, 76.6%) had a 5-year local recurrence-free survival of 85.6% (95% CI: 77.6-93.6%), (P = 0.003). According to the result of multivariable analysis, patients with lower NEO1 expression had a 2.7-fold increased risk of local tumor recurrence (95% CI: 1.0-7.0, P = 0.043) compared to patients with higher NEO1 expression. CONCLUSIONS: HNSCC patients with a lower transcriptional expression of NEO1 have a significantly higher risk of local recurrence after surgical treatment.

6.
Cancers (Basel) ; 14(9)2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35565388

ABSTRACT

The expression of the semaphorin-3F (SEMA3F) and neuropilin-2 (NRP2) is involved in the regulation of lymphangiogenesis. The present study analyzes the relationship between the transcriptional expression of the SEMA3F-NRP2 genes and the presence of occult lymph node metastases in patients with cN0 head and neck squamous cell carcinomas. We analyzed the transcriptional expression of SEMA3F and NRP2 in a cohort of 53 patients with cN0 squamous cell carcinoma treated with an elective neck dissection. Occult lymph node metastases were found in 37.7% of the patients. Patients with occult lymph node metastases (cN0/pN+) had significantly lower SEMA3F expression values than patients without lymph node involvement (cN0/pN0). Considering the expression of the SEMA3F-NRP2 genes, patients were classified into two groups according to the risk of occult nodal metastasis: Group 1 (n = 34), high SEMA3F/low NRP2 expression, with a low risk of occult nodal involvement (14.7% cN0/pN+); Group 2 (n = 19), low SEMA3F or high SEMA3F/high NRP2 expression, with a high risk of occult nodal involvement (78.9% cN0/pN+). Multivariate analysis showed that patients in Group 2 had a 26.2 higher risk of lymph node involvement than patients in Group 1. There was a significant relationship between the transcriptional expression values of the SEMA3F-NRP2 genes and the risk of occult nodal metastases.

7.
Muscle Nerve ; 43(2): 268-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21254094

ABSTRACT

Proteolysis in skeletal muscle is mainly carried out by the activity of the ubiquitin-dependent proteolytic system. For the study of protein degradation through the ubiquitin-proteasome pathway, we used a model of hyperthermia in murine myotubes. In C2C12 cells, hyperthermia (41°C) induced a significant increase in both the rate of protein synthesis (18%) and degradation (51%). Interestingly, the addition of the ß(2) -adrenoceptor agonist formoterol resulted in a significant decrease in protein degradation (21%) without affecting protein synthesis. The decrease in proteolytic rate was associated with decreases in gene expression of the different components of the ubiquitin-dependent proteolytic system. The effects of the ß(2) -agonist on protein degradation were dependent exclusively on cAMP formation, because inhibition of adenylyl cyclase completely abolished the effects of formoterol on protein degradation. It can be concluded that hyperthermia is a suitable model for studying the anti-proteolytic potential of drugs used in the treatment of muscle wasting.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Ethanolamines/pharmacology , Hyperthermia, Induced , Muscle Fibers, Skeletal/drug effects , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism , Analysis of Variance , Animals , Cell Line, Transformed , Cyclic AMP/metabolism , DDT/analogs & derivatives , DDT/pharmacology , Formoterol Fumarate , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Immunosuppressive Agents/pharmacology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myofibrils/metabolism , Proteasome Endopeptidase Complex/genetics , Ubiquitins/genetics
8.
Crit Care ; 15(3): R124, 2011.
Article in English | MEDLINE | ID: mdl-21569477

ABSTRACT

INTRODUCTION: Survivors of critical illness often have significant long-term brain dysfunction, and routine clinical procedures like mechanical ventilation (MV) may affect long-term brain outcome. We aimed to investigate the effect of the increase of tidal volume (Vt) on brain activation in a rat model. METHODS: Male Sprague Dawley rats were randomized to three groups: 1) Basal: anesthetized unventilated animals, 2) low Vt (LVt): MV for three hours with Vt 8 ml/kg and zero positive end-expiratory pressure (ZEEP), and 3) high Vt (HVt) MV for three hours with Vt 30 ml/kg and ZEEP. We measured lung mechanics, mean arterial pressure (MAP), arterial blood gases, and plasma and lung levels of cytokines. We used immunohistochemistry to examine c-fos as a marker of neuronal activation. An additional group of spontaneously breathing rats was added to discriminate the effect of surgical procedure and anesthesia in the brain. RESULTS: After three hours on LVt, PaO2 decreased and PaCO2 increased significantly. MAP and compliance remained stable in MV groups. Systemic and pulmonary inflammation was higher in MV rats than in unventilated rats. Plasma TNFα was significantly higher in HVt than in LVt. Immunopositive cells to c-fos in the retrosplenial cortex and thalamus increased significantly in HVt rats but not in LVt or unventilated rats. CONCLUSIONS: MV promoted brain activation. The intensity of the response was higher in HVt animals, suggesting an iatrogenic effect of MV on the brain. These findings suggest that this novel cross-talking mechanism between the lung and the brain should be explored in patients undergoing MV.


Subject(s)
Brain Injuries/physiopathology , Brain/physiology , Respiration, Artificial/adverse effects , Tidal Volume/physiology , Animals , Biomarkers/analysis , Disease Models, Animal , Lung/physiology , Male , Proto-Oncogene Proteins c-fos/analysis , Random Allocation , Rats , Rats, Sprague-Dawley
9.
Cancers (Basel) ; 13(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201840

ABSTRACT

Breast cancer (BrCa) is the leading cause of death among women worldwide, with about one million new cases diagnosed each year. In spite of the improvements in diagnosis, early detection and treatment, there is still a high incidence of mortality and failure to respond to current therapies. With the use of several well-established biomarkers, such as hormone receptors and human epidermal growth factor receptor-2 (HER2), as well as genetic analysis, BrCa patients can be categorized into multiple subgroups: Luminal A, Luminal B, HER2-enriched, and Basal-like, with specific treatment strategies. Although chemotherapy and targeted therapies have greatly improved the survival of patients with BrCa, there is still a large number of patients who relapse or who fail to respond. The role of the tumor microenvironment in BrCa progression is becoming increasingly understood. Cancer-associated fibroblasts (CAFs) are the principal population of stromal cells in breast tumors. In this review, we discuss the current understanding of CAFs' role in altering the tumor response to therapeutic agents as well as in fostering metastasis in BrCa. In addition, we also review the available CAFs-directed molecular therapies and their potential implications for BrCa management.

10.
Muscle Nerve ; 42(6): 936-49, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21104868

ABSTRACT

The hypothesis we tested was that administering corticotropin-releasing factor receptor agonists preserves muscle mass during cancer that is related to changes in tissue gene expression. cDNA microarrays were used to compare mRNAs from muscle and adipose tissues of non-treated and agonist-treated tumor-bearing rats. In muscle of non-tumor-bearing agonist-treated animals we observed decreased expression of genes associated with fatty acid uptake and esterification. In tumor-bearing animals, CRF2R agonist administration produced decreased mRNA content of the atrogene lipin-1. In white adipose tissue, agonist treatment of non-tumor-bearing animals induced genes typically related to muscle structure and function. The fact that this treatment decreased expression of atrogenes could have clinical application. In addition, agonist treatment changed the gene pattern of adipose tissue to render it similar to that of skeletal muscle; thus, treatment with this agonist alters the gene pattern to what could be called "muscularization of white adipose tissue."


Subject(s)
Adipose Tissue/metabolism , Cachexia/metabolism , Corticotropin-Releasing Hormone/pharmacology , Muscle, Skeletal/metabolism , Receptors, Corticotropin-Releasing Hormone/agonists , Adipose Tissue/drug effects , Analysis of Variance , Animals , Cachexia/genetics , Corticotropin-Releasing Hormone/metabolism , Gene Expression , Male , Muscle, Skeletal/drug effects , Neoplasm Transplantation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
12.
Curr Opin Otolaryngol Head Neck Surg ; 28(2): 107-111, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32022731

ABSTRACT

PURPOSE OF REVIEW: A strong association between diabetes mellitus and carcinogenesis has been reported in different organs. The purpose of this review is to summarize the new evidences in relation to diabetes mellitus and its association with the development, prognosis, and therapeutic strategies of head and neck squamous cell carcinomas (HNSCC). RECENT FINDINGS: Recent publications suggest that glycemic metabolism is altered in HNSCC. Elevated blood glucose levels, before or around the time of diagnosis, have been reported to reduce survival rates in HNSCC. Also, the homeostasis model assessment-insulin resistance has been independently associated with disease-free survival, suggesting that improving the glycemic control may improve the prognosis in this group of patients.Epidemiological studies revealed that cancer patients with diabetes mellitus have less cancer-related mortality after antiglycemic treatment, opening the option to include antiglycolytic agents, such as metformin, in the therapeutic plan. This finding is in accordance with in-vitro studies that demonstrated a decrease in tumor-cell proliferation with antidiabetic medications. SUMMARY: Recent findings highlight the importance of glucose metabolism in the pathogenesis and progression of cancer cells. The knowledge of these altered pathways gives us an opportunity to design target treatments aimed to modulate glucose catabolism.


Subject(s)
Diabetes Mellitus/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/therapy , Blood Glucose/metabolism , Disease Progression , Humans , Prognosis , Risk Factors
13.
J Vis Exp ; (163)2020 09 24.
Article in English | MEDLINE | ID: mdl-33044448

ABSTRACT

Tumorigenesis is a multi-step process in which cells acquire capabilities that allow their growth, survival, and dissemination under hostile conditions. Different tests seek to identify and quantify these hallmarks of cancerous cells; however, they often focus on a single aspect of cellular transformation and, in fact, multiple tests are required for their proper characterization. The purpose of this work is to provide researchers with a set of tools to assess cellular transformation in vitro from a broad perspective, thereby making it possible to draw sound conclusions. A sustained proliferative signaling activation is the major feature of tumoral tissues and can be easily monitored under in vitro conditions by calculating the number of population doublings achieved over time. Besides, the growth of cells in 3D cultures allows their interaction with surrounding cells, resembling what occurs in vivo. This enables the evaluation of cellular aggregation and, together with immunofluorescent labeling of distinctive cellular markers, to obtain information on another relevant feature of tumoral transformation: the loss of proper organization. Another remarkable characteristic of transformed cells is their capacity to grow without attachment to other cells and to the extracellular matrix, which can be evaluated with the anchorage assay. Detailed experimental procedures to evaluate cell growth rate, to perform immunofluorescent labeling of cell lineage markers in 3D cultures, and to test anchorage-independent cell growth in soft agar are provided. These methodologies are optimized for Breast Primary Epithelial Cells (BPEC) due to its relevance in breast cancer; however, procedures can be applied to other cell types after some adjustments.


Subject(s)
Breast/pathology , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Animals , Basement Membrane/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Culture Techniques , Cell Polarity , Cell Proliferation , Cells, Cultured , Female , Fluorescent Antibody Technique , Humans , Image Processing, Computer-Assisted , Models, Biological , Signal Transduction , Software
14.
J Clin Med ; 9(3)2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32178425

ABSTRACT

Current evidences state clear that both normal development of breast tissue as well as its malignant progression need many-sided local and systemic communications between epithelial cells and stromal components. During development, the stroma, through remarkably regulated contextual signals, affects the fate of the different mammary cells regarding their specification and differentiation. Likewise, the stroma can generate tumour environments that facilitate the neoplastic growth of the breast carcinoma. Mammographic density has been described as a risk factor in the development of breast cancer and is ascribed to modifications in the composition of breast tissue, including both stromal and glandular compartments. Thus, stroma composition can dramatically affect the progression of breast cancer but also its early detection since it is mainly responsible for the differences in mammographic density among individuals. This review highlights both the pathological and biological evidences for a pivotal role of the breast stroma in mammographic density, with particular emphasis on dense and malignant stromas, their clinical meaning and potential therapeutic implications for breast cancer patients.

15.
Clin Cancer Res ; 26(6): 1432-1448, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31699826

ABSTRACT

PURPOSE: Despite the therapeutic success of existing HER2-targeted therapies, tumors invariably relapse. This study aimed at identifying new mechanisms responsible for HER2-targeted therapy resistance. EXPERIMENTAL DESIGN: We have used a platform of HER2-targeted therapy-resistant cell lines and primary cultures of healthy and tumor-associated fibroblasts (TAF) to identify new potential targets related to tumor escape from anti-HER2 therapies. RESULTS: We have shown that TAFs promote resistance to HER2-targeted therapies. TAFs produce and secrete high levels of FGF5, which induces FGFR2 activation in the surrounding breast cancer cells. FGFR2 transactivates HER2 via c-Src, leading to resistance to HER2-targeted therapies. In vivo, coinoculating nonresistant cell lines with TAFs results in more aggressive and resistant tumors. Resistant cells activate fibroblasts and secrete FGFR ligands, creating a positive feedback loop that fuels resistance. FGFR2 inhibition not only inhibits HER2 activation, but also induces apoptosis in cells resistant to HER2-targeted therapies. In vivo, inhibitors of FGFR2 reverse resistance and resensitize resistant cells to HER2-targeted therapies. In HER2 patients' samples, α-SMA, FGF5, and FGFR2 contribute to poor outcome and correlate with c-Src activation. Importantly, expression of FGF5 and phospho-HER2 correlated with a reduced pathologic complete response rate in patients with HER2-positive breast cancer treated with neoadjuvant trastuzumab, which highlights the significant role of TAFs/FGF5 in HER2 breast cancer progression and resistance. CONCLUSIONS: We have identified the TAF/FGF5/FGFR2/c-Src/HER2 axis as an escape pathway responsible for HER2-targeted therapy resistance in breast cancer, which can be reversed by FGFR inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cancer-Associated Fibroblasts/pathology , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Female , Humans , Lapatinib/administration & dosage , Mice , Mice, Nude , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Receptor, ErbB-2/metabolism , Signal Transduction , Survival Rate , Trastuzumab/administration & dosage , Xenograft Model Antitumor Assays
16.
Cancer Res ; 80(2): 276-290, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31694906

ABSTRACT

The tumor-promoting fibrotic stroma rich in tumor-associated fibroblasts (TAF) is drawing increased therapeutic attention. Intriguingly, a trial with the antifibrotic drug nintedanib in non-small cell lung cancer reported clinical benefits in adenocarcinoma (ADC) but not squamous cell carcinoma (SCC), even though the stroma is fibrotic in both histotypes. Likewise, we reported that nintedanib inhibited the tumor-promoting fibrotic phenotype of TAFs selectively in ADC. Here we show that tumor fibrosis is actually higher in ADC-TAFs than SCC-TAFs in vitro and patient samples. Mechanistically, the reduced fibrosis and nintedanib response of SCC-TAFs was associated with increased promoter methylation of the profibrotic TGFß transcription factor SMAD3 compared with ADC-TAFs, which elicited a compensatory increase in TGFß1/SMAD2 activation. Consistently, forcing global DNA demethylation of SCC-TAFs with 5-AZA rescued TGFß1/SMAD3 activation, whereas genetic downregulation of SMAD3 in ADC-TAFs and control fibroblasts increased TGFß1/SMAD2 activation, and reduced their fibrotic phenotype and antitumor responses to nintedanib in vitro and in vivo. Our results also support that smoking and/or the anatomic location of SCC in the proximal airways, which are more exposed to cigarette smoke particles, may prime SCC-TAFs to stronger SMAD3 epigenetic repression, because cigarette smoke condensate selectively increased SMAD3 promoter methylation. Our results unveil that the histotype-specific regulation of tumor fibrosis in lung cancer is mediated through differential SMAD3 promoter methylation in TAFs and provide new mechanistic insights on the selective poor response of SCC-TAFs to nintedanib. Moreover, our findings support that patients with ADC may be more responsive to antifibrotic drugs targeting their stromal TGFß1/SMAD3 activation. SIGNIFICANCE: This study implicates the selective epigenetic repression of SMAD3 in SCC-TAFs in the clinical failure of nintedanib in SCC and supports that patients with ADC may benefit from antifibrotic drugs targeting stromal TGFß1/SMAD3.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , Indoles/pharmacology , Lung Neoplasms/drug therapy , Smad3 Protein/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/surgery , Aged , Aged, 80 and over , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cohort Studies , DNA Methylation/genetics , Epigenetic Repression , Female , Fibrosis , Gene Expression Regulation, Neoplastic , Humans , Indoles/therapeutic use , Lung/cytology , Lung/drug effects , Lung/pathology , Lung/surgery , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Mice , Middle Aged , Pneumonectomy , Promoter Regions, Genetic/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Tissue Array Analysis , Xenograft Model Antitumor Assays
17.
Int J Mol Med ; 24(1): 63-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19513536

ABSTRACT

C2C12 cells exposed to hyperthermia (41 degrees C) experienced an increase in both protein synthesis and degradation. The addition of IL15 under hyperthermic conditions resulted in an important increase in protein synthesis with no changes in protein degradation, except when cells overexpressed PPARdelta. The PPARdelta agonist GW501516 exerted similar effects on protein synthesis to IL15. Expression of a mutant dominant negative form of PPARdelta prevented the effect of the cytokine on protein synthesis, suggesting that this transcription factor is involved in the anabolic action of IL15. The present study also suggests that the effects of IL15 on lipid oxidation could be mediated by PPARdelta.


Subject(s)
Hot Temperature , Interleukin-15/physiology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , PPAR delta/metabolism , Animals , Cell Line , Interleukin-15/pharmacology , Mice , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Mutation , PPAR delta/antagonists & inhibitors , PPAR delta/genetics , Protein Biosynthesis , Thiazoles/pharmacology
18.
Cancer Res ; 67(13): 6512-9, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616713

ABSTRACT

Implantation of the Yoshida AH-130 ascites hepatoma to rats resulted in a decrease in muscle weight 7 days after the inoculation of the tumor. These changes were associated with increases in the mRNA content for both peroxisome proliferator-activated receptor (PPAR) gamma and PPAR delta in skeletal muscle. The increase in gene expression for these transcription factors was related to increases in the expression of several genes involved in fatty acid transport, activation, and oxidation. Tumor burden also resulted in increases in PPAR gamma coactivator-1 alpha gene expression and pyruvate dehydrogenase kinase 4. All these changes in lipid metabolism genes suggest that a metabolic shift occurs in skeletal muscle of tumor-bearing rats toward a more oxidative phenotype. Formoterol treatment to tumor-bearing rats resulted in an amelioration of all the changes observed as a result of tumor burden. Administration of this beta(2)-adrenergic agonist also resulted in a decrease in mRNA content of muscle PPAR alpha, PPAR delta, and PPAR gamma, as well as in mRNA levels of many of the genes involved in both lipid and mitochondrial metabolism. All these results suggest an involvement of the different PPARs as transcription factors related with muscle wasting and also indicate that a possible mode of action of the anticachectic compound formoterol may involve a normalization of the levels of these transcription factors.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Gene Expression Regulation, Neoplastic , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Body Weight , Cachexia , Fatty Acids/metabolism , Lipid Metabolism , Male , Muscle, Skeletal/pathology , Muscular Atrophy , Neoplasms, Experimental , Rats , Rats, Wistar , Transcription, Genetic
19.
Cancer Lett ; 424: 70-83, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29548821

ABSTRACT

Histamine receptor 1 (HRH1) belongs to the rhodopsin-like G-protein-coupled receptor family. Its activation by histamine triggers cell proliferation, embryonic development, and tumor growth. We recently established that HRH1 is up-regulated in basal and human epidermal growth factor receptor 2 (HER2)-enriched human breast tumors and that its expression correlates with a worse prognosis. Nevertheless, the functional role of HRH1 in basal and HER2-targeted therapy-resistant breast cancer (BC) progression has not yet been addressed. Using terfenadine, a selective chemical inhibitor of HRH1, we showed that the inhibition of HRH1 activity in basal BC cells leads to sub-G0 cell accumulation, suppresses proliferation, promotes cell motility and triggers the activation of extracellular signal-regulated kinase (ERK) signaling, initiating the mitochondrial apoptotic pathway. Furthermore, HER2-targeted therapy-resistant cells express higher levels of HRH1 and are more sensitive to terfenadine treatment. Moreover, in vivo experiments showed that terfenadine therapy reduced the tumor growth of basal and trastuzumab-resistant BC cells. In conclusion, our results suggest that targeting HRH1 is a promising new clinical approach to consider that could enhance the effectiveness of current therapeutic treatment in patients with basal and BC tumors resistant to HER2-targeted therapies.


Subject(s)
Breast Neoplasms/drug therapy , Histamine H1 Antagonists, Non-Sedating/administration & dosage , MAP Kinase Signaling System/drug effects , Receptors, Histamine H1/metabolism , Terfenadine/administration & dosage , Trastuzumab/administration & dosage , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Histamine H1 Antagonists, Non-Sedating/pharmacology , Humans , MCF-7 Cells , Mice , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/metabolism , Receptor, ErbB-2/metabolism , Terfenadine/pharmacology , Trastuzumab/pharmacology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
20.
Biochim Biophys Acta ; 1761(1): 37-42, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16458591

ABSTRACT

Interleukin 15 (IL-15) has previously been shown to have important effects on lipid metabolism in adipose tissue, particularly influencing the rate of the de novo fatty acid synthesis. The results presented here show that chronic administration to rats (100 microg/kg body weight) has important effects on the metabolic fate of an exogenous [(14)C]-triolein load, decreasing the incorporation of lipid into adipose tissue and significantly increasing the total (14)CO(2) formation from [(14)C]-triolein. Skeletal muscle and possibly liver seem to be the main organs involved in the action of IL-15 on lipid oxidation, since the presence of the cytokine in incubated EDL muscle with [(14)C]-palmitic acid increased (14)CO(2) formation by 39%. Concerning the mechanism, the results suggest that the transport of fatty acids into mitochondria could be involved in the action of IL-15 since the cytokine clearly increases the presence of L-CPT-I and CPT-II in liver tissue. In addition, IL-15 treatment resulted in a significant increment in the gene expression of PPARdelta, a transcription factor clearly related with lipid catabolism in many tissues. Altogether, the results presented here suggest that IL-15 alters exogenous lipid partitioning, limiting adipose tissue uptake and favouring oxidation.


Subject(s)
Interleukin-15/pharmacology , Triolein/metabolism , Animals , Interleukin-15/genetics , Male , Organ Specificity , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptors/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL