Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Cells ; 13(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38920644

ABSTRACT

Hepatocellular carcinoma (HCC) development is associated with altered modifications in DNA methylation, changing transcriptional regulation. Emerging evidence indicates that DNA methyltransferase 1 (DNMT1) plays a key role in the carcinogenesis process. This study aimed to investigate how pirfenidone (PFD) modifies this pathway and the effect generated by the association between c-Myc expression and DNMT1 activation. Rats F344 were used for HCC development using 50 mg/kg of diethylnitrosamine (DEN) and 25 mg/kg of 2-Acetylaminofluorene (2-AAF). The HCC/PFD group received simultaneous doses of 300 mg/kg of PFD. All treatments lasted 12 weeks. On the other hand, HepG2 cells were used to evaluate the effects of PFD in restoring DNA methylation in the presence of the inhibitor 5-Aza. Histopathological, biochemical, immunohistochemical, and western blot analysis were carried out and our findings showed that PFD treatment reduced the amount and size of tumors along with decreased Glipican-3, ß-catenin, and c-Myc expression in nuclear fractions. Also, this treatment improved lipid metabolism by modulating PPARγ and SREBP1 signaling. Interestingly, PFD augmented DNMT1 and DNMT3a protein expression, which restores global methylation, both in our in vivo and in vitro models. In conclusion, our results suggest that PFD could slow down HCC development by controlling DNA methylation.


Subject(s)
Carcinoma, Hepatocellular , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Proliferating Cell Nuclear Antigen , Pyridones , Animals , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Pyridones/pharmacology , Rats , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Humans , Hep G2 Cells , Proliferating Cell Nuclear Antigen/metabolism , Male , Rats, Inbred F344 , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Diethylnitrosamine , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/genetics
2.
Curr Issues Mol Biol ; 46(4): 3050-3062, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38666921

ABSTRACT

Nearly 90% of oral cancers are characterized as oral squamous cell carcinoma (OSCC), representing the sixth most common type of cancer. OSCC usually evolves from oral potentially malignant disorders that, in some cases, are histologically consistent with a oral dysplasia. The levels of 1α,25 dihydroxyvitamin D3 (1,25-(OH)2D3; calcitriol), the active form of vitamin D3, have been shown to be decreased in patients with oral dysplasia and OSCC. Moreover, treatment with 1,25-(OH)2D3 has been proven beneficial in OSCC by inhibiting the Wnt/ß-catenin pathway, a signaling route that promotes cell migration, proliferation, and viability. However, whether this inhibition mechanism occurs in oral dysplasia is unknown. To approach this question, we used dysplastic oral keratinocyte cultures and oral explants (ex vivo model of oral dysplasia) treated with 1,25-(OH)2D3 for 48 h. Following treatment with 1,25-(OH)2D3, both in vitro and ex vivo models of oral dysplasia showed decreased levels of nuclear ß-catenin by immunofluorescence (IF) and immunohistochemistry (IHC). Consistently, reduced protein and mRNA levels of the Wnt/ß-catenin target gene survivin were observed after treatment with 1,25-(OH)2D3. Moreover, 1,25-(OH)2D3 promoted membranous localization of E-cadherin and nuclear localization of vitamin D receptor (VDR). Functionally, DOK cells treated with 1,25-(OH)2D3 displayed diminished cell migration and viability in vitro.

3.
Cir Cir ; 2024 03 19.
Article in English | MEDLINE | ID: mdl-38502938

ABSTRACT

Objective: Estrogen (E2) plays a significant role in postmenopausal osteoporosis, and its deficiency is related to chronic low-grade inflammation. Intravenous immunoglobulin (IVIG) is composed of immunoglobulins derived from the plasma of healthy donors. Numerous anti-inflammatory pathways are responsible for IVIG's anti-inflammatory action The aim of this study is to investigate the effects of IVIG on experimental-induced osteoporosis. Materials and methods: Forty adult female Wistar rats were included in the study. Thirty rats underwent bilateral dorsal ovariectomy. Rats were grouped as Group 1 (n = 10, ovariectomy and saline); Group 2 (n = 10, ovariectomy and E2); Group 3 (n = 10, ovariectomy and IVIG), and Control group (n = 10, no oophorectomy). Histopathological examination of bone tissue, and biochemical analysis for beta-catenin, plasma Tumor Necrosis Factor-α, IL-6, receptor activator of nuclear-κB ligand (RANKL), and osteoprotegerin (OPG) levels were made. Results: The IVIG group had increased trabecular number, area, and thickness with increased bone mineral density as well as decreased trabecular separation compared with the saline group. IVIG group had lower serum RANKL and higher serum OPG levels when compared with the saline group. The bone marrow beta-catenin level was significantly higher in the control and ovariectomy + IVIG groups. Conclusion: IVIG has beneficial effects on experimentally induced osteoporosis with a possible action on inflammation and RANKL-ß-catenin pathway.


Objetivo: El estrógeno juega un papel importante en la osteoporosis posmenopáusica y su deficiencia está relacionada con la inflamación crónica de bajo grado. La inmunoglobulina intravenosa (IGIV) está compuesta por inmunoglobulinas derivadas del plasma de donantes sanos. El objetivo de este estudio es investigar los efectos de IVIG en la osteoporosis inducida experimentalmente. Materiales y métodos: 30 ratas se sometieron a ovariectomía dorsal bilateral. las ratas se agruparon como: Grupo 1 (n = 10, ovariectomía y solución salina); Grupo 2 (n = 10, ovariectomía y estrógeno); Grupo 3 (n = 10, ovariectomía e IVIG) y Grupo Control (n = 10, sin ovariectomía). Se realizó un examen histopatológico del tejido óseo y un análisis bioquímico de los niveles de beta-catenina, factor de necrosis tumoral α (TNF-α), IL-6, RANKL y osteoprotegerina (OPG) en plasma. Resultados: El grupo IVIG había aumentado el número, el área y el grosor trabecular con una mayor densidad mineral ósea, así como una menor separación trabecular en comparación con el grupo de solución salina. El nivel de beta-catenina en la médula ósea fue significativamente mayor en los grupos de control y de ovariectomía + IVIG. Conclusión: IVIG tiene efectos beneficiosos sobre la osteoporosis inducida experimentalmente con una posible acción sobre la inflamación y la vía RANKL-ß-catenina.

4.
J Cell Commun Signal ; 17(4): 1389-1403, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37982965

ABSTRACT

Aberrant canonical Wnt signaling is a hallmark of colon cancer. The TP53 tumor suppressor gene is altered in many solid tumors, including colorectal cancer, resulting in mutant versions of p53 (mut-p53) that lose their tumor suppressor capacities and acquire new-oncogenic functions (GOFs) critical for disease progression. Although the mechanisms related to mut-p53 GOF have been explored extensively, the relevance of mut-p53 in the canonical Wnt pathway is not well defined. This work investigated the influence of mut-p53 compared to wt-p53 in ß-catenin-dependent Wnt signaling. Using the TCGA public data from Pan-Cancer and the GEPIA2 platform, an in silico analysis of wt-p53 versus mut-p53 genotyped colorectal cancer patients showed that TP53 (p53) and CTNNB1 (ß-catenin) are significantly overexpressed in colorectal cancer, compared with normal tissue. Using p53 overexpression or p53 knockdown assays of wt-p53 or mut-p53, we found that while wt-p53 antagonizes canonical Wnt signaling, mut-p53 induces the opposite effect, improving the ß-catenin-dependent transcriptional activity and colony formation ability of colon cancer cells, which were both decreased by mut-p53 knockdown expression. The mechanism involved in mut-p53-induced activation of canonical Wnt appears to be via AKT-mediated phosphorylation of Ser 552 of ß-catenin, which is known to stabilize and enhance its transcriptional activity. We also found that while wt-p53 expression contributes to 5-FU sensitivity in colon cancer cells, the RITA p53 reactivating molecule counteracted the resistance against 5-FU in cells expressing mut-p53. Our results indicate that mut-p53 GOF acts as a positive regulator of canonical Wnt signaling and participates in the induction of resistance to 5-FU in colon cancer cells.

5.
Gac Med Mex ; 159(3): 240-246, 2023.
Article in English | MEDLINE | ID: mdl-37494709

ABSTRACT

Colorectal cancer (CRC) is a complex disease, determined by genetic, environmental and lifestyle-associated risk factors. Genetic (inherited) factors have great influence on its development; however, most cases of CRC are sporadic and gradually develop over several years. The main environmental risk factors are associated with b-catenin signaling pathway, including obesity, lack of physical activity, consumption of red and processed meats, alcoholism, and smoking. The pathway is related to cell homeostasis regulation and cell self-renewal during embryogenesis and adulthood. The main recommendation for preventing the development of CRC is to reduce the risk factors, increase the consumption of fruits, vegetables and grains, exercise regularly and limit the consumption of both alcohol and tobacco. However, family history and the presence of a hereditary syndrome increase the risk, which is why carrying out periodic examinations to detect CRC is suggested, using development predictors such as biochemical and molecular markers, which are discussed in this work.


El cáncer colorrectal (CCR) es una enfermedad compleja determinada por factores de riesgo genéticos, ambientales y de estilo de vida. Los factores genéticos (hereditarios) tienen gran influencia en su desarrollo, sin embargo, la mayoría de los casos de CCR son esporádicos y se desarrollan gradualmente a lo largo de varios años. Los principales factores ambientales de riesgo están asociados a la vía de señalización de ß-catenina, entre ellos obesidad, falta de actividad física, consumo de carnes rojas y procesadas, alcoholismo y tabaquismo. La vía está relacionada con la regulación de la homeostasis celular, autorrenovación celular durante la embriogénesis y edad adulta. La principal recomendación para evitar el desarrollo del CCR es reducir los factores de riesgo, aumentar el consumo de frutas, verduras y granos, hacer ejercicio de manera rutinaria y limitar el consumo tanto de alcohol como de tabaco. Dado que los antecedentes familiares y la presencia de un síndrome hereditario aumentan el riesgo, se sugiere hacer exámenes periódicos para detectar CCR y emplear predictores del desarrollo como los marcadores bioquímicos y moleculares, los cuales se presentan en este trabajo.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , beta Catenin/metabolism , Risk Factors , Obesity/complications , Signal Transduction , Gene Expression Regulation, Neoplastic
6.
Pharmaceuticals (Basel) ; 16(2)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-37259421

ABSTRACT

Colorectal cancer mortality rate and highly altered proteins from the Wnt/ß-catenin pathway increase the scientific community's interest in finding alternatives for prevention and treatment. This study aims to determine the biological effect of chlorogenic acid (CGA) on two colorectal cancer cell lines, HT-29 and SW480, and its interactions with ß-catenin and LRP6 to elucidate a possible modulatory mechanism on the Wnt/ß-catenin pathway. These effects were determined by propidium iodide and DiOC6 for mitochondrial membrane permeability, MitoTracker Red for mitochondrial ROS production, DNA content for cell distribution on cell cycle phases, and molecular docking for protein-ligand interactions and binding affinity. Here, it was found that CGA at 2000 µM significantly affects cell viability and causes DNA fragmentation in SW480 cells rather than in HT-29 cells, but in both cell lines, it induces ROS production. Additionally, CGA has similar affinity and interactions for LRP6 as niclosamide but has a higher affinity for both ß-catenin sites than C2 and iCRT14. These results suggest a possible modulatory role of CGA over the Wnt/ß-catenin pathway in colorectal cancer.

7.
Clinics (Sao Paulo) ; 78: 100189, 2023.
Article in English | MEDLINE | ID: mdl-37015185

ABSTRACT

OBJECTIVE: To explore whether the effect of ß-catenin on MI and MI-induced cardiomyocyte apoptosis is YAP-dependent. METHODS: The authors established an MI rat model by ligating the anterior descending branch of the left coronary artery, and an MI cell model by treating cardiomyocytes with H2O2. RESULTS: ß-catenin downregulation was observed in MI cardiac tissues and in H2O2-treated cardiomyocytes. Lentiviral-CTNNB1 was administered to MI rats to upregulate ß-catenin expression in MI cardiac tissue. ß-catenin recovery reduced the myocardial infarct area, fibrosis, and apoptotic cell death in MI rats. H2O2 treatment attenuated cell viability and induced cell death in cardiomyocytes, whereas ß-catenin overexpression partially reversed these changes. Moreover, H2O2 treatment caused the deactivation of Yes-Associated Protein (YAP), as detected by increased YAP phosphorylation and reduced the nuclear localization of YAP. Upregulation of ß-catenin expression reactivated YAP in H2O2-treated cardiomyocytes. Reactivation of YAP was achieved by administration of Mitochonic Acid-5 (MA-5) to H2O2-treated cardiomyocytes, and deactivation of YAP by CIL56 treatment in ß-catenin-overexpressing H2O2-treated cardiomyocytes. MA-5 administration increased cell viability and repressed apoptosis in H2O2-treated cardiomyocytes, whereas CIL56 treatment counteracted the effects of ß-catenin overexpression on cell survival and apoptosis. CONCLUSIONS: The present data indicate that ß-catenin and YAP are effective treatment targets for MI, blocking the apoptotic death of cardiomyocytes.


Subject(s)
Myocardial Infarction , beta Catenin , Animals , Rats , Apoptosis , beta Catenin/metabolism , Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/metabolism
8.
Front Endocrinol (Lausanne) ; 14: 1124111, 2023.
Article in English | MEDLINE | ID: mdl-36936148

ABSTRACT

Prostate cancer remains the most prevalent cancer among men worldwide. This cancer is hormone-dependent; therefore, androgen, estrogen, and their receptors play an important role in development and progression of this disease, and in emergence of the castration-resistant prostate cancer (CRPC). Galectins are a family of ß-galactoside-binding proteins which are frequently altered (upregulated or downregulated) in a wide range of tumors, participating in different stages of tumor development and progression, but the molecular mechanisms which regulate its expression are still poorly understood. This review provides an overview of the current and emerging knowledge on Galectin-3 in cancer biology with focus on prostate cancer and the interplay with estrogen receptor (ER) signaling pathways, present in androgen-independent prostate cancer cells. We suggest a molecular mechanism where ER, Galectin-3 and ß-catenin can modulate nuclear transcriptional events, such as, proliferation, migration, invasion, and anchorage-independent growth of androgen-independent prostate cancer cells. Despite a number of achievements in targeted therapy for prostate cancer, CRPC may eventually develop, therefore new effective drug targets need urgently to be found. Further understanding of the role of Galectin-3 and ER in prostate cancer will enhance our understanding of the molecular mechanisms of prostate cancer development and the future treatment of this disease.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Estrogen , Galectin 3/genetics , Androgens/therapeutic use , Receptors, Androgen/metabolism , Galectins
10.
World J Gastroenterol ; 28(26): 3027-3046, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-36051330

ABSTRACT

Colorectal cancer (CRC) continues to be one of the main causes of death from cancer because patients progress unfavorably due to resistance to current therapies. Dysregulation of the Wnt/ß-catenin pathway plays a fundamental role in the genesis and progression of several types of cancer, including CRC. In many subtypes of CRC, hyperactivation of the ß-catenin pathway is associated with mutations of the adenomatous polyposis coli gene. However, it can also be associated with other causes. In recent years, studies of the tumor microenvironment (TME) have demonstrated its importance in the development and progression of CRC. In this tumor nest, several cell types, structures, and biomolecules interact with neoplastic cells to pave the way for the spread of the disease. Cross-communications between tumor cells and the TME are then established primarily through paracrine factors, which trigger the activation of numerous signaling pathways. Crucial advances in the field of oncology have been made in the last decade. This Minireview aims to actualize what is known about the central role of the Wnt/ß-catenin pathway in CRC chemoresistance and aggressiveness, focusing on cross-communication between CRC cells and the TME. Through this analysis, our main objective was to increase the understanding of this complex disease considering a more global context. Since many treatments for advanced CRC fail due to mechanisms involving chemoresistance, the data here exposed and analyzed are of great interest for the development of novel and effective therapies.


Subject(s)
Colorectal Neoplasms , beta Catenin , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Tumor Microenvironment , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
11.
Front Endocrinol (Lausanne) ; 13: 870172, 2022.
Article in English | MEDLINE | ID: mdl-35928898

ABSTRACT

Clinically silent corticotroph tumors are usually macroadenomas that comprise 20% of ACTH tumors. They frequently progress to aggressive tumors with high recurrence, invasiveness, and on rare occasions, they may become hormonally active causing Cushing's disease. Trustable biomarkers that can predict their aggressive course, as well as their response to traditional or new therapies, are paramount. Aberrant ß-Catenin expression and localization have been proposed as responsible for several malignancies including pituitary tumors. Nevertheless, the role of ß-Catenin in the aggressive transformation of silent corticotropinomas and their response to Temozolomide salvage treatment have not been explored yet. In this work, we present a case of a silent corticotroph tumor that invaded cavernous sinus and compressed optic chiasm and, after a first total resection and tumor remission it recurred six years later as an aggressive ACTH-secreting tumor. This lesion grew with carotid compromise and caused Cushing's signs. It required multiple medical treatments including Cabergoline, Ketoconazole, TMZ, and radiotherapy. Besides, other two surgeries were needed until it could be controlled. Interestingly, we found α-SMA vascular area reduction and differential ß-Catenin cell localization in the more aggressive tumor stages characterized by high Ki-67 indexes and p53 expression. Our results may indicate a role of angiogenesis and ß-Catenin trigged events in the pituitary tumor progression, which could in turn affect the response to TMZ and/or conventional treatments. These molecular findings in this unusual case could be useful for future management of aggressive pituitary tumors.


Subject(s)
Adenoma , Pituitary Neoplasms , Adenoma/pathology , Adrenocorticotropic Hormone/metabolism , Corticotrophs/metabolism , Corticotrophs/pathology , Humans , Pituitary Neoplasms/pathology , Temozolomide/therapeutic use , beta Catenin/metabolism
12.
Cancer Biol Ther ; 23(1): 1-13, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35944058

ABSTRACT

The PI3K/Akt and Wnt/ß-catenin pathways play an important role in the acquisition of the malignant phenotype in cancer. However, there are few data regarding the role of the interplay between both pathways in colorectal cancer (CRC) progression. The mutational status and the clinicopathological characteristics of PI3K/Akt and Wnt/ß-catenin pathways were accessed by bioinformatic analysis whereas that the impact of the interplay between the activity of both pathways to explain tumorigenic potential was performed in vitro using IGF-1 and Wnt3a treatments in CRC cell models. The mutational status of these pathways did not influence the survival of CRC patients, but an association between clinicopathological characteristics in patients with mutations in one, but not in both pathways was observed. A potentiating effect on the activation of both pathways and enhanced cellular migration and proliferation was observed when both pathways were activated simultaneously with IGF-1 and Wnt3a. In addition, these effects were hindered after pretreatment with LY294002, a specific PI3K inhibitor, suggesting some dependence between these two signaling cascades. Our findings show that, regardless of mutational status, there is an interplay between the activity of PI3K/Akt and Wnt/ß-catenin pathways that contributes to events related to CRC progression and that the reversal of such events using a PI3K inhibitor highlights the value of targeting these pathways for potential directed therapies in CRC patients.


Subject(s)
Colorectal Neoplasms , beta Catenin , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/pharmacology , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
13.
Tumour Biol ; 44(1): 85-105, 2022.
Article in English | MEDLINE | ID: mdl-35811548

ABSTRACT

INTRODUCTION: Prolactinomas are the most frequent pituitary tumor subtype. Despite most of them respond to medical treatment, a proportion are resistant and become a challenge in clinical management. Wnt/ß-Catenin pathway has been implicated in several cancers including pituitary tumors and other sellar region malignancies. Interestingly, Wnt/ß-Catenin inhibition augments the cytotoxicity of the chemotherapeutic agent Temozolomide (TMZ) in different cancers. TMZ is now being implemented as rescue therapy for aggressive pituitary adenoma treatment. However, the molecular mechanisms associated with TMZ action in pituitary tumors remain unclear. OBJECTIVES: Our aims in the present study were to evaluate differential ß-Catenin expression in human resistant prolactinomas and Wnt/ß-Catenin signaling activation and involvement in Prolactin (PRL) secreting experimental models treated with TMZ. RESULTS: We first evaluated by immunohistochemistry ß-Catenin localization in human resistant prolactinomas in which we demonstrated reduced membrane ß-Catenin in prolactinoma cells compared to normal pituitaries, independently of the Ki-67 proliferation indexes. In turn, in vivo 15 mg/kg of orally administered TMZ markedly reduced PRL production and increased prolactinoma cell apoptosis in mice bearing xenografted prolactinomas. Intratumoral ß-Catenin strongly correlated with Prl and Cyclin D1, and importantly, TMZ downregulated both ß-Catenin and Cyclin D1, supporting their significance in prolactinoma growth and as candidates of therapeutic targets. When tested in vitro, TMZ directly reduced MMQ cell viability, increased apoptosis and produced G2/M cell cycle arrest. Remarkably, ß-Catenin activation and VEGF secretion were inhibited by TMZ in vitro. CONCLUSIONS: We concluded that dopamine resistant prolactinomas undergo a ß-Catenin relocalization in relation to normal pituitaries and that TMZ restrains experimental prolactinoma tumorigenicity by reducing PRL production and ß-Catenin activation. Together, our findings contribute to the understanding of Wnt/ß-Catenin implication in prolactinoma maintenance and TMZ therapy, opening the opportunity of new treatment strategies for aggressive and resistant pituitary tumors.


Subject(s)
Pituitary Neoplasms , Prolactinoma , Animals , Cyclin D1 , Humans , Mice , Models, Theoretical , Pituitary Neoplasms/pathology , Prolactin/metabolism , Prolactin/therapeutic use , Prolactinoma/drug therapy , Prolactinoma/metabolism , Prolactinoma/pathology , Temozolomide/pharmacology , Temozolomide/therapeutic use , beta Catenin
14.
Int J Mol Sci ; 24(1)2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36613445

ABSTRACT

Recently, the combination of chemotherapy plus nivolumab (chemo-immunotherapy) has become the standard of care for advanced-stage gastric cancer (GC) patients. However, despite its efficacy, up to 40% of patients do not respond to these treatments. Our study sought to identify variations in gene expression associated with primary resistance to chemo-immunotherapy. Diagnostic endoscopic biopsies were retrospectively obtained from advanced GC patients previously categorized as responders (R) or non-responders (NR). Thirty-four tumor biopsies (R: n = 16, NR: n = 18) were analyzed by 3' massive analysis of cDNA ends (3'MACE). We found >30 differentially expressed genes between R and NRs. Subsequent pathway enrichment analyses demonstrated that angiogenesis and the Wnt-ß-catenin signaling pathway were enriched in NRs. Concomitantly, we performed next generation sequencing (NGS) analyses in a subset of four NR patients that confirmed alterations in genes that belonged to the Wnt/ß-catenin and the phosphoinositide 3-kinase (PI3K) pathways. We speculate that angiogenesis, the Wnt, and the PI3K pathways might offer actionable targets. We also discuss therapeutic alternatives for chemo-immunotherapy-resistant advanced-stage GC patients.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , beta Catenin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Retrospective Studies , Wnt Signaling Pathway/genetics , Immunotherapy , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
15.
Mol Cell Endocrinol ; 538: 111454, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34520813

ABSTRACT

Hypothyroidism is a protective factor against breast cancer but long-term exposure or overdoses of thyroid replacement therapy with thyroxine (T4) may increase breast cancer risk. OBJECTIVE: to study, in vivo and in vitro, the effects of T4 on the proliferation and apoptosis of mammary tumors of hypo- and euthyroid rats, and the possible mechanisms involved in these effects. MATERIAL AND METHODS: Female Sprague-Dawley rats were treated with a single dose of dimethylbenzathracene (15 mg/rat) at 55 days of age and were divided into three groups: hypothyroidism (HypoT; 0.01% 6-N-propyl-2-thiouracil -PTU- in drinking water, n = 20), hypothyroidism treated with T4 (HypoT + T4; 0.01% PTU in drinking water and 0.25 mg/kg/day T4 via sc; n = 20) and EUT (untreated control, n = 20). At sacrifice, tumor explants from HypoT and EUT rats were obtained and treated either with 10-10 M T4 in DMEM/F12 without phenol red with 1% Charcoalized Fetal Bovine Serum or DMEM/F12 only for 15 min to evaluate intracellular signaling pathways associated with T4, and 24 h to evaluate changes in the expression of hormone receptors and proteins related to apoptosis and proliferation by immunohistochemistry and Western Blot. RESULTS: In vivo, hypothyroidism retards mammary carcinogenesis but its treatment with T4 reverted the protective effects. In vitro, the proliferative and anti-apoptosis mechanisms of T4 were different regarding the thyroid status. In EUT tumors, the main signaling pathway involved was the cross-talk with other receptors, such as ERα, PgR, and HER2. In HypoT tumors, the non-genomic signaling pathway of T4 was the chief mechanism involved since αvß3 integrin, HER2, ß-catenin and, downstream, PI3K/AKT and ERK signaling pathways were activated. CONCLUSION: T4 can regulate mammary carcinogenesis by mainly activating its non-genomic signaling pathway and by interacting with other hormone or growth factor pathways endorsing that overdoses of thyroid replacement therapy with T4 can increase the risk of breast cancer.


Subject(s)
Anthracenes/adverse effects , Hypothyroidism/drug therapy , Mammary Neoplasms, Experimental/metabolism , Piperidines/adverse effects , Propylthiouracil/adverse effects , Signal Transduction/drug effects , Thyroxine/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hypothyroidism/chemically induced , Mammary Neoplasms, Experimental/chemically induced , Rats , Rats, Sprague-Dawley , Thyroxine/pharmacology
16.
Pharmaceuticals (Basel) ; 14(9)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34577571

ABSTRACT

The Wnt/ß-catenin signaling pathway plays a major role in cell survival and proliferation, as well as in angiogenesis, migration, invasion, metastasis, and stem cell renewal in various cancer types. However, the modulation (either up- or downregulation) of this pathway can inhibit cell proliferation and apoptosis both through ß-catenin-dependent and independent mechanisms, and by crosstalk with other signaling pathways in a wide range of malignant tumors. Existing studies have reported conflicting results, indicating that the Wnt signaling can have both oncogenic and tumor-suppressing roles, depending on the cellular context. This review summarizes the available information on the role of the Wnt/ß-catenin pathway and its crosstalk with other signaling pathways in apoptosis induction in cancer cells and presents a modified dual-signal model for the function of ß-catenin. Understanding the proapoptotic mechanisms induced by the Wnt/ß-catenin pathway could open new therapeutic opportunities.

17.
Clin Transl Oncol ; 23(12): 2448-2459, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34426910

ABSTRACT

Aberrant Wnt/ß-catenin signaling is central to colorectal cancer carcinogenesis. The well-known potential of targeting the canonical Wnt signaling pathway for the treatment of CRC is largely attributed to the ability of this pathway to regulate various cellular processes such as cell proliferation, metastasis, drug resistance, immune response, apoptosis, and cellular metabolism. However, with the current approach of targeting this pathway, none of the Wnt-targeted agents have been successfully implicated in clinical practice. Instead of using classical approaches to target this pathway, there is a growing need to find new and modified approaches to achieve the same. For this, a better understanding of the regulation of ß-catenin, a major effector of the canonical Wnt pathway is a must. The present review addresses the importance of understanding the regulation of ß-catenin beyond the destruction complex. Few recently discovered ß-catenin regulators such as ZNF281, TTPAL, AGR2, ARHGAP25, TREM2, and TIPE1 showed significant potential in regulating the development of CRC through modulation of the Wnt/ß-catenin signaling pathway in both in vitro and in vivo studies. Although the expression and activity of ß-catenin is influenced by many protein regulators, the abovementioned proteins not only influence its expression and activation but are also directly involved in the development of CRC and various other solid tumors. Therefore, we hypothesise that focusing the current research on finding the detailed mechanism of action of these regulators may assist in providing with a better treatment approach or improve the current therapeutic regimens.


Subject(s)
Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Carcinogenesis/metabolism , Humans , Neoplasms/metabolism
18.
Reprod Toxicol ; 103: 139-148, 2021 08.
Article in English | MEDLINE | ID: mdl-34146661

ABSTRACT

Bisphenols are a group of environmental endocrine-disrupting chemicals that produce alterations in the expression of intercellular junction proteins of the Blood-Testis Barrier (BTB) involved in spermatogenesis. The association between bisphenol exposure and BTB protein expression is controversial. Therefore, we performed this systematic review and meta-analysis to clarify bisphenol effects on Sertoli cell BTB protein expression in vitro. The Standardized Mean Difference (SMD) with a 95 % confidence interval (95 % CI) was used to evaluate the association between alterations in the BTB protein expression and bisphenol exposure in vitro. Six articles were included in the meta-analysis. Bisphenol-A (BPA) exposure at 200 µM was associated with significant decrease in BTB protein expression (SMD = -2.70, 95 %CI: -3.59, -1.80, p het = 0.46, p = <0.00001). In the moderate (40-50 µM) and low dose (<25 µM), no significant associations were obtained. We also found a non-monotonic dose-response curve of bisphenol effect in ZO-1 protein expression; low and high doses presented a significant decrease compared to control, while moderate dose presented no change. The current temporary Tolerable Daily Intake (tTDI) of BPA is 4 µg/kg bw/day. The 5-25 µM doses of BPA are equivalent to ∼1-5 mg/kg bw, respectively. Although the low dose group (<25 µM) assessed doses below the previous NOAEL value, these doses are above the current tTDI. Thus, it is necessary to conduct more studies with lower bisphenol concentrations to avoid underestimating the potential adverse effects of bisphenols at doses below tTDI.


Subject(s)
Benzhydryl Compounds/toxicity , Blood-Testis Barrier/drug effects , Phenols/toxicity , Endocrine Disruptors/metabolism , Endocrine Disruptors/toxicity , Humans , Intercellular Junctions/drug effects , Male , Occludin/metabolism , Proteins/metabolism , Sertoli Cells/drug effects , Spermatogenesis , Testis/drug effects , Zonula Occludens-1 Protein/metabolism
19.
Food Chem Toxicol ; 153: 112263, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34015426

ABSTRACT

In this study, the changes in oncogenic and tumor suppressor signaling pathways in liver and their association with serum and urinary biomarkers of aflatoxin exposure were evaluated in Wistar rats fed diets containing aflatoxin B1 (AFB1) for 90 days. Rats were divided into four groups (n = 15 per group) and assigned to dietary treatments containing 0 (control), 50 (AFB50), 100 (AFB100) and 200 µg AFB1 kg-1 diet (AFB200). Multiple preneoplastic foci of hepatocytes marked with glutathione-S-transferase-placental form (GST-P) were identified in AFB100 and AFB200 groups. Hepatocellular damage induced by AFB1 resulted in overexpression of cyclin D1 and ß-catenin. The liver expression of retinoblastoma (Rb) and p27Kip1 decreased in AFB100 and AFB200 groups, confirming the favorable conditions for neoplastic progression to hepatocellular carcinoma. All samples from rats fed AFB1-contaminated diets had quantifiable AFB1-lysine in serum or urinary AFM1 and AFB1-N7-guanine, with mean levels of 20.42-50.34 ng mL-1, 5.31-37.68 and 39.15-126.37 ng mg-1 creatinine, respectively. Positive correlations were found between AFB1-lysine, AFM1 or AFB1-N7-guanine and GST-P+, ß-catenin+ and cyclin D1+ hepatocytes, while Rb + cells negatively correlated with those AFB1 exposure biomarkers. The pathways evaluated are critical molecular mechanisms of AFB1-induced hepatocarcinogenesis in rats.


Subject(s)
Aflatoxin B1/toxicity , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Retinoblastoma Protein/metabolism , beta Catenin/metabolism , Aflatoxin B1/analogs & derivatives , Aflatoxin B1/blood , Aflatoxin B1/metabolism , Aflatoxin B1/urine , Aflatoxin M1/urine , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Biomarkers/blood , Biomarkers/urine , Gene Expression/drug effects , Guanine/analogs & derivatives , Guanine/urine , Hepatocytes/drug effects , Liver/drug effects , Liver/pathology , Lysine/blood , Male , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Rats, Wistar
20.
Int J Mol Sci ; 22(3)2021 Jan 25.
Article in English | MEDLINE | ID: mdl-33503805

ABSTRACT

Castration-resistant prostate cancer (CRPC) is an advanced and androgen-independent form of prostate cancer. Recent studies of rapid actions mediated by estrogen in the prostate and its relationship with CRPC are emerging. We have previously shown that estrogen receptor (ER) promotes migration and invasion of the androgen-independent prostate cancer cells PC-3, but the signaling pathways involved in these events remain to be elucidated. Therefore, this study aimed to analyze the role of ERα and ERß in the activation of SRC, and the involvement of SRC and PI3K/AKT on invasion and colony formation of the PC-3 cells. Our results showed that the activation of ERα (using ERα-selective agonist PPT) and ERß (using ERß-selective agonist DPN) increased phosphorylation of SRC in PC-3 cells. In the presence of the selective inhibitor for SRC-family kinases PP2, the effects of DPN and PPT on transmigration and soft agar colony formation assays were decreased. Furthermore, SRC is involved in the expression of the non-phosphorylated ß-catenin. Finally, using PI3K specific inhibitor Wortmannin and AKT inhibitor MK2206, we showed that PI3K/AKT are also required for invasion and colony formation of PC-3 cells simulated by ER. This study provides novel insights into molecular mechanisms of ER in PC-3 cells by demonstrating that ER, located outside the cell nucleus, activates rapid responses molecules, including SRC and PI3K/AKT, which enhance the tumorigenic potential of prostate cancer cells, increasing cell proliferation, migration, invasion, and tumor formation.


Subject(s)
Androgens/metabolism , Prostatic Neoplasms/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Humans , Immunohistochemistry , Male , PC-3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL