Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Clin Transl Oncol ; 23(8): 1542-1548, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33948919

ABSTRACT

BACKGROUND: Cancerous inhibitor of protein phosphatase 2A (CIP2A) plays a critical role in the pathogenesis of various types of cancer. Here, we investigated whether manipulating CIP2A abundance could enhance the treatment effects of doxorubicin in MCF-7/ADR cells. METHODS: CIP2A silencing was achieved by specific siRNAs. Proliferation of breast cancer cell line MCF-7/ADR under effective doxorubicin concentrations after CIP2A silencing was examined by MTT assay. Wound healing assay was performed to quantify cell migration and caspase-3/-7 activities were measured for assessing the extent of apoptosis. RESULTS: First, our data confirmed that MCF-7/ADR cell proliferation was suppressed by doxorubicin in a dose-dependent manner. Additionally, knocking down of CIP2A could further decrease MCF-7 cell proliferation and migration, even in the presence of doxorubicin. Mechanistically, we have found that CIP2A silencing promoted cell apoptosis relative to doxorubicin alone or vehicle control groups. Lastly, phosphatase2A (PP2A) activity was potentiated and the autophagy markers, LC3B and Beclin1, were upregulated after knocking down CIP2A. CONCLUSION: Our findings support the potential benefits of using CIP2A inhibitor as a therapeutic agent to treat doxorubicin-resistant breast cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Autoantigens/genetics , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Silencing , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Protein Phosphatase 2/metabolism , Apoptosis , Autoantigens/physiology , Autophagy , Beclin-1/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Movement , Cell Proliferation/drug effects , Female , Humans , Intracellular Signaling Peptides and Proteins/physiology , MCF-7 Cells , Membrane Proteins/physiology , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering , Up-Regulation
2.
J Alzheimers Dis ; 82(s1): S163-S178, 2021.
Article in English | MEDLINE | ID: mdl-33612542

ABSTRACT

BACKGROUND: Disturbances in the autophagy/endolysosomal systems are proposed as early signatures of Alzheimer's disease (AD). However, few studies are available concerning autophagy gene expression in AD patients. OBJECTIVE: To explore the differential expression of classical genes involved in the autophagy pathway, among them a less characterized one, DEF8 (Differentially expressed in FDCP 8), initially considered a Rubicon family member, in peripheralblood mononuclear cells (PBMCs) from individuals with mild cognitive impairment (MCI) and probable AD (pAD) and correlate the results with the expression of DEF8 in the brain of 5xFAD mice. METHOD: By real-time PCR and flow cytometry, we evaluated autophagy genes levels in PBMCs from MCI and pAD patients. We evaluated DEF8 levels and its localization in brain samples of the 5xFAD mice by real-time PCR, western blot, and immunofluorescence. RESULTS: Transcriptional levels of DEF8 were significantly reduced in PBMCs of MCI and pAD patients compared with healthy donors, correlating with the MoCA and MoCA-MIS cognitive tests scores. DEF8 protein levels were increased in lymphocytes from MCI but not pAD, compared to controls. In the case of brain samples from 5xFAD mice, we observed a reduced mRNA expression and augmented protein levels in 5xFAD compared to age-matched wild-type mice. DEF8 presented a neuronal localization. CONCLUSION: DEF8, a protein proposed to act at the final step of the autophagy/endolysosomal pathway, is differentially expressed in PBMCs of MCI and pAD and neurons of 5xFAD mice. These results suggest a potential role for DEF8 in the pathophysiology of AD.


Subject(s)
Alzheimer Disease/metabolism , Autophagy/physiology , Brain/metabolism , Cognitive Dysfunction/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Biomarkers/metabolism , Brain/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Transgenic , Middle Aged
3.
Curr Mol Med ; 21(1): 2-4, 2021.
Article in English | MEDLINE | ID: mdl-32660402

ABSTRACT

The COVID-19 plague is hitting mankind. Several viruses, including SARS-CoV-1, MERS-CoV, EBOV, and SARS-CoV-2, use the endocytic machinery to enter the cell. Genomic variants in NPC1, which encodes for the endo-lysosomal Niemann-Pick type C1 protein, restricts the host-range of viruses in bats and susceptibility to infections in humans. Lack of NPC1 and its pharmacological suppression inhibits many viral infections including SARS-CoV-1 and Type I Feline Coronavirus Infection. Antiviral effects of NPC1-inhibiting drugs for COVID-19 treatment should be explored.


Subject(s)
COVID-19/physiopathology , Intracellular Signaling Peptides and Proteins/physiology , SARS-CoV-2/physiology , Antiviral Agents/therapeutic use , COVID-19/virology , Drug Repositioning , Humans , Niemann-Pick C1 Protein , Severity of Illness Index , Virus Internalization , COVID-19 Drug Treatment
4.
Basic Clin Pharmacol Toxicol ; 114(6): 476-84, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24345272

ABSTRACT

Renal thioredoxin reductase-1 (TrxR-1) activity is stimulated at lead doses lower than that necessary to inhibit δ-aminolevulinate dehydratase activity (δ-ALA-D), which is a classical early biomarker of lead effects. Thus, we hypothesized that the activity of TrxR-1 could be a more sensitive early indicator of lead effects than is δ-ALA-D. To evaluate this hypothesis, we assessed the blood and renal TrxR-1 activity and its gene expression along with biomarkers of oxidative damage, antioxidant enzyme activities and biomarkers of lead exposure in rats acutely exposed to lead. A histopathological analysis was performed to verify renal damage. The increase in renal TrxR-1 activity paralleled the increase in the blood and renal lead levels at 6, 24 and 48 hr after the exposure to 25 mg/kg lead acetate (p < 0.05), whereas its expression was increased 24 and 48 hr after exposure. These effects were not accompanied by oxidative or tissue damage in the kidneys. Blood TrxR-1 activity was not affected by lead exposure (up to 25 mg/kg). Erythrocyte δ-ALA-D activity was inhibited 6 hr after the exposure to 25 mg/kg lead acetate (p < 0.05) but recovered thereafter. Renal δ-ALA-D activity decreased 24 and 48 hr after the exposure to 25 mg/kg lead acetate. There were no changes in any parameters at lead acetate doses <25 mg/kg. Our results indicate that blood TrxR-1 activity is not a suitable indicator of lead effects. In contrast, the increase in renal TrxR-1 expression and activity is implicated in the early events of lead exposure, most likely as a protective cellular mechanism against lead toxicity.


Subject(s)
Cytosol/enzymology , Kidney/drug effects , Lead/toxicity , Thioredoxin Reductase 1/metabolism , Animals , Erythrocytes/enzymology , Intracellular Signaling Peptides and Proteins/physiology , Kelch-Like ECH-Associated Protein 1 , Kidney/enzymology , Kidney/pathology , Lead/pharmacokinetics , Male , Porphobilinogen Synthase/metabolism , Rats , Rats, Wistar , Thioredoxin Reductase 1/genetics
5.
Regul Pept ; 188: 25-30, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24333629

ABSTRACT

AIM: Orexin A and orexin B (hypocretins) are neuropeptides synthesized mainly by neurons located in the lateral hypothalamus and projections throughout the brain. They are agonists at both the orexin 1 and orexin 2G protein-coupled receptors. They have been related to arousal, sleep and feeding, autonomic and neuroendocrine functions. Their role in the brain control of gonadotropins secretion was postulated in rodents and humans. Previously, we demonstrated the participation of the orexinergic system in attaining successful reproduction in in vivo studies. METHODS: We studied in vitro the effects of both neuropeptides, in the presence or absence of selective antagonists, on the mRNA expression of orexin 1 and orexin 2 receptors in anterior pituitary cells of proestrous rats, as well as the direct effects on FSH and LH secretion. RESULTS: Both orexin A and orexin B increased FSH and LH secretion; these effects were suppressed by the orexin 1 receptor blocking agent SB-334867 and the orexin 2 receptor antagonists JNJ-10397049. Orexin A and orexin B decreased OX1 receptor mRNA expression and this effect was modified only when both blocking agents were present. Neither orexin A nor the blocking drugs by themselves modified OX2 receptor mRNA expression. Orexin B treatment increased the mRNA expression of OX2 receptor. The effect was abolished only by the OX2 receptor antagonist. CONCLUSION: In an in vitro model, we demonstrated a direct effect of orexins on gonadotropins release and orexins receptors expression, underlining the hypothesis that orexins participate in the brain control of pituitary functions.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Orexin Receptors/metabolism , Pituitary Gland, Anterior/cytology , Animals , Cells, Cultured , Estrous Cycle , Female , Follicle Stimulating Hormone/metabolism , Gene Expression , Luteinizing Hormone/metabolism , Orexin Receptors/genetics , Orexins , Rats , Rats, Sprague-Dawley
6.
Invest Clin ; 55(4): 371-91, 2014 Dec.
Article in Spanish | MEDLINE | ID: mdl-25558756

ABSTRACT

Surgery, radiotherapy and chemotherapy are universally recognized as the most effective anti-cancer therapies. Despite significant advances directed towards elucidating molecular mechanisms and developing clinical trials, cancer still remains a major public health issue. Cancer stem cells are a subpopulation of the cells that form the tumor. The discovery of these human cancer cells opens a perspective for understanding tumor recurrence, drug resistance and metastasis; and opens up new research directions on how cancer cells are capable of switching from dormancy to malignancy. Therapeutic alternatives emerge from a better understanding of the biology and the environment of tumor stem cells. The present paper aims to summarize the characteristics and properties of cancer stem cells, the ongoing research, as well as the best strategies for prevention and control of the mechanisms of tumor recurrence.


Subject(s)
Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Cell Differentiation , Cell Transformation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Models, Biological , Neoplasm Metastasis , Neoplasm Proteins/physiology , Neoplasm Recurrence, Local , Neoplasms/therapy , Neoplastic Cells, Circulating , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Signal Transduction/physiology , Stem Cells/classification , Stem Cells/cytology
7.
Invest Clin ; 54(2): 186-205, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23947008

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a biological phenomenon responsible for the formation of different tissues and organs during normal metazoan development. Because of the connection of the EMT with the pathogenesis of certain diseases, such as cancer, the attention of the scientific community has been directed towards the search for and identification of effective therapeutic targets. These targets include signal transduction in cancerous stem cells and the use of microRNAs, which would inhibit EMT-associated phenotypic changes and tumoral progression. In an attempt to compile relevant and current information, this work addresses concepts that define the EMT and the advances in this field. The wealth of knowledge gained from areas such as the loss of cell polarity and intracellular adhesion complexes, the signaling pathways implicated, microRNA participation in this process, and stemness acquisition in embryonic and cancerous cells, all of which allow for the visualization of promising perspectives, particularly, methods for targeting advanced malignancies, are presented herein.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition , Molecular Targeted Therapy , Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Adhesion , Cell Adhesion Molecules/physiology , Cell Differentiation , Cell Movement , Cell Polarity , Cell Transformation, Neoplastic , Disease Progression , Embryonic Development , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Fibrosis , Humans , Intracellular Signaling Peptides and Proteins/physiology , MicroRNAs/physiology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , RNA, Neoplasm/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Invest. clín ; Invest. clín;54(2): 186-205, jun. 2013. ilus
Article in English | LILACS | ID: lil-740348

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a biological phenomenon responsible for the formation of different tissues and organs during normal metazoan development. Because of the connection of the EMT with the pathogenesis of certain diseases, such as cancer, the attention of the scientific community has been directed towards the search for and identification of effective therapeutic targets. These targets include signal transduction in cancerous stem cells and the use of microRNAs, which would inhibit EMT-associated phenotypic changes and tumoral progression. In an attempt to compile relevant and current information, this work addresses concepts that define the EMT and the advances in this field. The wealth of knowledge gained from areas such as the loss of cell polarity and intracellular adhesion complexes, the signaling pathways implicated, microRNA participation in this process, and stemness acquisition in embryonic and cancerous cells, all of which allow for the visualization of promising perspectives, particularly, methods for targeting advanced malignancies, are presented herein.


La transición epitelio-mesenquimática (TEM) es el fenómeno biológico responsable de la formación de los diferentes tejidos y órganos durante el desarrollo normal de los organismos metazoarios. En razón de su conexión con la patogénesis de ciertas enfermedades como el cáncer, la atención de la comunidad científica se ha redireccionado hacia la búsqueda e identificación de blancos terapéuticos efectivos, como la transducción de señales de las células madre cancerosas o la utilización de microARNs, que permitirían bloquear los cambios fenotípicos asociados con la TEM y, por ende, la progresión tumoral. En un intento por recopilar información relevante y actualizada, el presente trabajo aborda conceptos que definen a la TEM y avances alcanzados en este campo. El acervo de conocimiento obtenido en aspectos como pérdida de la polaridad celular y de los complejos de adhesión intercelular, vías de señalización implicadas y participación de los microARNs en el proceso, así como adquisición de stemness o troncalidad, tanto en células embrionarias como cancerosas, hace posible visualizar perspectivas promisorias, en especial en lo que se refiere a las terapias contra las malignidades de alto grado.


Subject(s)
Animals , Humans , Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition , Molecular Targeted Therapy , Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Polarity , Cell Transformation, Neoplastic , Cell Adhesion Molecules/physiology , Disease Progression , Embryonic Development , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Fibrosis , Intracellular Signaling Peptides and Proteins/physiology , MicroRNAs/physiology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , RNA, Neoplasm/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Mol Cell Biochem ; 378(1-2): 117-26, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23459830

ABSTRACT

ING proteins are tumor suppressors involved in the regulation of gene transcription, cell cycle arrest, apoptosis, and senescence. Here, we show that ING1b expression is upregulated by several DNA-damaging agents, in a p53-independent manner. ING1b stimulates DNA repair of a variety of DNA lesions requiring activation of multiple DNA repair pathways. Moreover, Ing1(-/-) cells showed impaired genomic DNA repair after H2O2 and neocarzinostatin treatment and this defect was reverted by overexpression of ING1b. Two tumor-derived ING1 mutants failed to promote DNA repair highlighting the physiological importance of the integrity of the PHD domain for ING1b DNA repair activity and suggesting a role in the prevention of tumor progression. Ing(-/-) cells showed higher basal levels of γ-H2AX and, upon DNA damage, γ-H2AX increase was greater and with faster kinetics compared to wild-type cells. Chromatin relaxation by Trichostatin A led to an exacerbated damage signal in both types of cells, but this effect was dependent on Ing1 status, and more pronounced in wild-type cells. Our results suggest that ING1 acts at early stages of the DNA damage response activating a variety of repair mechanisms and that this function of ING1 is targeted in tumors.


Subject(s)
DNA Repair , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , Gene Expression , Genome, Human , Genomic Instability , Histones/metabolism , Humans , Inhibitor of Growth Protein 1 , Mice , Mutation, Missense , Protein Isoforms/physiology , Protein Kinases/metabolism , Up-Regulation
10.
Brain Res ; 1491: 68-77, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23122879

ABSTRACT

Within the postero-lateral hypothalamus neurons that utilize hypocretin or melanin-concentrating hormone (MCH) as neuromodulators are co-distributed. These neurons have been involved in the control of behavioral states, and a deficit in the hypocretinergic system is the pathogenic basis of narcolepsy with cataplexy. In this report, utilizing immunohistochemistry and retrograde tracing techniques, we examined the hypocretinergic innervation of the nucleus pontis oralis (NPO), which is the executive site that is responsible for the generation of REM sleep in the cat. The retrograde tracer cholera toxin subunit b (CTb) was administered in pontine regions where carbachol microinjections induced REM sleep. Utilizing immunohistochemical techniques, we found that approximately 1% of hypocretinergic neurons in the tuberal area of the hypothalamus project to the NPO. In addition, approximately 6% of all CTb+ neurons in this region were hypocretinergic. The hypocretinergic innervation of the NPO was also compared with the innervation of the same site by MCH-containing neurons. More than three times as many MCHergic neurons were found to project to the NPO compared with hypocretinergic cells; both neuronal types exhibited bilateral projections. We also identified a group of non-hypocretinergic non-MCHergic neuronal group of neurons that were intermingled with both hypocretinergic and MCHergic neurons that also projected to this same brainstem region. These neurons were grater in number that either hypocretin or MCH-containing neurons; their soma size was also smaller and their projections were mainly ipsilateral. The present anatomical data suggest that hypocretinergic, MCHergic and an unidentified companion group of neurons of the postero-lateral hypothalamus participate in the regulation of the neuronal activity of NPO neurons, and therefore, are likely to participate in the control of wakefulness and REM sleep.


Subject(s)
Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neural Pathways/physiology , Neuropeptides/physiology , Pons/physiology , Sleep, REM/physiology , Animals , Carbachol/pharmacology , Cats , Cholera Toxin , Hypothalamic Hormones/pharmacology , Immunohistochemistry , Male , Melanins/pharmacology , Microinjections , Neural Pathways/drug effects , Orexins , Pituitary Hormones/pharmacology , Polysomnography , Sleep, REM/drug effects
11.
Regul Pept ; 178(1-3): 56-63, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22749989

ABSTRACT

Orexin-A and orexin-B are neuropeptides controlling sleep-wakefulness, feeding and neuroendocrine functions via their G protein-coupled receptors, orexin-1R and orexin-2R. They are synthesized in the lateral hypothalamus and project throughout the brain. Orexins and orexin receptors have also been described outside the brain. Previously we demonstrated the presence of both receptors in the ovary, their increased expression during proestrous afternoon and the dependence on the gonadotropins. Here we studied the effects of orexins on the mRNA expression of both receptors, by quantitative real-time PCR, on luteal cells from superovulated rat ovaries and granulosa cells from diethylstilbestrol-treated rat ovaries. Effects on progesterone secretion were also measured. In luteal cells, 1 nM of either orexin-A or orexin-B decreased progesterone secretion. Orexin-A treatment increased expression of both orexin-1R and orexin-2R mRNA. The effect on orexin-1R mRNA expression was abolished by an orexin-1R selective receptor antagonist SB-334867 and the effect on orexin-2R mRNA expression was abolished by a selective orexin-2R antagonist JNJ-10397049. Orexin-B did not modify orexin-1R mRNA expression, but increased orexin-2R mRNA expression. The effect of orexin-B on orexin-2R was abolished by a selective orexin-2R antagonist. Neither the expression of orexin receptors nor progesterone secretions by granulosa cells were affected by orexins. FSH, as positive control, increased both steroid hormones secretion, but did not induce the expression of OX receptors in granulosa cells isolated from late preantral/early antral follicles. Finally in ovaries obtained immediately after sacrifice, the expression of orexin-1R and orexin-2R was higher in superovulated rat ovaries compared to control or diethylstilbestrol treated rat ovaries. A selective presence and function of both orexinergic receptors in luteal and granulosa cells is described, suggesting that the orexinergic system may have a functional role in the ovary.


Subject(s)
Granulosa Cells/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Luteal Cells/metabolism , Neuropeptides/physiology , Progesterone/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Benzoxazoles/pharmacology , Cells, Cultured , Diethylstilbestrol/pharmacology , Dioxanes/pharmacology , Estradiol/metabolism , Estrogens, Non-Steroidal/pharmacology , Female , Gene Expression , Naphthyridines , Orexin Receptors , Orexins , Ovary/cytology , Ovary/metabolism , Phenylurea Compounds/pharmacology , Progesterone/blood , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/genetics , Urea/analogs & derivatives , Urea/pharmacology
12.
Plant Signal Behav ; 7(1): 53-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22301969

ABSTRACT

The recent finding of a tissue-specific cell cycle regulator (SCI1) that inhibits cell proliferation/differentiation in the upper pistil points to an unanticipated way of controlling plant morphogenesis. The similarity between the SCI1 RNAi-silenced plants and some auxin-related phenotypes suggested that SCI1 could be involved in the auxin signaling pathway. To address this hypothesis, we analyzed the expression of three auxin-related genes in transgenic plants in which SCI1 was silenced and overexpressed. The results showed that the expression levels of the auxin-related genes largely correlated with the SCI1 expression level. Additionally, we analyzed the Arabidopsis SCI1 upstream regulatory region and found putative cis-acting elements also present in the AtCYCB1;1 AtYUC1, AtYUC2 and AtYUC4 URRs, suggesting a cell cycle- and auxin-related transcriptional regulation. Based on our previous and the current studies, we propose SCI1 as a signal transducer engaging auxin signaling and cell division/differentiation.


Subject(s)
Arabidopsis Proteins/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Indoleacetic Acids/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Signal Transduction
13.
Cir Cir ; 80(4): 389-98, 2012.
Article in Spanish | MEDLINE | ID: mdl-23374390

ABSTRACT

The Wnt-ß-catenin signalling pathway plays a crucial role in the regulation, differentiation, proliferation and cellular death processes; consequently, alterations in this pathway are involved in numerous abnormalities of development, growth and homeostasis in animal organisms. Wnt proteins include a numerous family of secretion glycoproteins which join to Frizzled receptors and Low Density Lipoprotein Receptor-related Protein, in order to stabilize the critical ß-catenin protein, and to initiate an intricate signaling cascade, which is related to multiple nucleocytoplasmatic processes. Alterations in the canonical Wnt-ß-catenin signaling pathway have been associated with variations in a number of proteins participating in this route, or with activation / inactivation of oncogenes and tumor suppressor genes, which explain different processes of tumorigenesis, in addition to a number of malformations and human diseases. This review describes the relations between the Wnt-ß-catenin signaling pathway with different neoplasic processes, as well as its application in the diagnosis and prognosis of cancer.


Subject(s)
Neoplasms/physiopathology , Wnt Signaling Pathway , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Genetic Therapy , Humans , Intracellular Signaling Peptides and Proteins/physiology , Leukemia/genetics , Leukemia/metabolism , Leukemia/physiopathology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Wnt Proteins/physiology , beta Catenin/physiology
14.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;44(4): 276-282, Apr. 2011. ilus, tab
Article in English | LILACS | ID: lil-581490

ABSTRACT

The signaling lymphocytic activation molecule (SLAM), present on the surface of hematopoietic cells, can regulate some events of the immune responses. This modulatory action is associated with the capacity of SLAM to interact with an intracytoplasmic adapter, such as SLAM-associated protein (SAP). SLAM is constitutively expressed in most of these cells, is rapidly induced after antigenic or inflammatory stimuli, and participates in the immunological synapse. Defects in the function of the SLAM-SAP pathway contribute to immunological abnormalities, resulting in autoimmune diseases, tumors of the lymphoid tissues and inadequate responses to infectious agents. Initially, the role of SLAM was investigated using an anti-SLAM monoclonal antibody (α-SLAM mAb) identified as an agonist of the SLAM-SAP pathway, which could induce the production of interferon-γ and could redirect the immune response to a T helper 1 (Th1) cell profile. However, in this review we postulate that the SLAM-SAP pathway primarily induces a Th2 response and secondarily suppresses the Th1 response.


Subject(s)
Humans , Antigens, CD/physiology , /metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lymphocyte Activation , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Antibodies, Monoclonal , Th1 Cells/immunology , Th1 Cells/metabolism , /immunology , /metabolism
15.
Braz J Med Biol Res ; 44(4): 276-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21445533

ABSTRACT

The signaling lymphocytic activation molecule (SLAM), present on the surface of hematopoietic cells, can regulate some events of the immune responses. This modulatory action is associated with the capacity of SLAM to interact with an intracytoplasmic adapter, such as SLAM-associated protein (SAP). SLAM is constitutively expressed in most of these cells, is rapidly induced after antigenic or inflammatory stimuli, and participates in the immunological synapse. Defects in the function of the SLAM-SAP pathway contribute to immunological abnormalities, resulting in autoimmune diseases, tumors of the lymphoid tissues and inadequate responses to infectious agents. Initially, the role of SLAM was investigated using an anti-SLAM monoclonal antibody (α-SLAM mAb) identified as an agonist of the SLAM-SAP pathway, which could induce the production of interferon-γ and could redirect the immune response to a T helper 1 (Th1) cell profile. However, in this review we postulate that the SLAM-SAP pathway primarily induces a Th2 response and secondarily suppresses the Th1 response.


Subject(s)
Antigens, CD/physiology , CD4-Positive T-Lymphocytes/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lymphocyte Activation , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Antibodies, Monoclonal , Humans , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family Member 1 , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
16.
J Biol Chem ; 285(31): 24131-40, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20495004

ABSTRACT

Target of rapamycin (TOR) kinases are highly conserved protein kinases that integrate signals from nutrients and growth factors to coordinate cell growth and cell cycle progression. It has been previously described that two TOR kinases control cell growth in the protozoan parasite Trypanosoma brucei, the causative agent of African trypanosomiasis. Here we studied an unusual TOR-like protein named TbTOR-like 1 containing a PDZ domain and found exclusively in kinetoplastids. TbTOR-like 1 localizes to unique cytosolic granules. After hyperosmotic stress, the localization of the protein shifts to the cell periphery, different from other organelle markers. Ablation of TbTOR-like 1 causes a progressive inhibition of cell proliferation, producing parasites accumulating in the S/G(2) phase of the cell cycle. TbTOR-like 1 knocked down cells have an increased area occupied by acidic vacuoles, known as acidocalcisomes, and are enriched in polyphosphate and pyrophosphate. These results suggest that TbTOR-like 1 might be involved in the control of acidocalcisome and polyphosphate metabolism in T. brucei.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Polyphosphates/chemistry , Protein Serine-Threonine Kinases/physiology , Trypanosoma brucei brucei/metabolism , Animals , Cell Cycle , Cell Proliferation , Cytosol/metabolism , DNA, Kinetoplast/metabolism , Diphosphates/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Models, Biological , Organelles/metabolism , Osmosis , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Protozoan Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
17.
J Endocrinol ; 206(1): 113-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20392814

ABSTRACT

Phosphoinositide-3-kinase (PI3K) inhibition increases functional sodium iodide symporter (NIS) expression in both FRTL-5 rat thyroid cell line and papillary thyroid cancer lineages. In several cell types, the stimulation of PI3K results in downstream activation of the mechanistic target of rapamycin (MTOR), a serine-threonine protein kinase that is a critical regulator of cellular metabolism, growth, and proliferation. MTOR activation is involved in the regulation of thyrocyte proliferation by TSH. Here, we show that MTOR inhibition by rapamycin increases iodide uptake in TSH-stimulated PCCL3 thyroid cell line, although the effect of rapamycin was less pronounced than PI3K inhibition. Thus, NIS inhibitory pathways stimulated by PI3K might also involve the activation of proteins other than MTOR. Insulin downregulates iodide uptake and NIS protein expression even in the presence of TSH, and both effects are counterbalanced by MTOR inhibition. NIS protein expression levels were correlated with iodide uptake ability, except in cells treated with TSH in the absence of insulin, in which rapamycin significantly increased iodide uptake, while NIS protein levels remained unchanged. Rapamycin avoids the activation of both p70 S6 and AKT kinases by TSH, suggesting the involvement of MTORC1 and MTORC2 in TSH effect. A synthetic analog of rapamycin (everolimus), which is clinically used as an anticancer agent, was able to increase rat thyroid iodide uptake in vivo. In conclusion, we show that MTOR kinase participates in the control of thyroid iodide uptake, demonstrating that MTOR not only regulates cell survival, but also normal thyroid cell function both in vitro and in vivo.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Sodium Iodide/metabolism , Thyroid Gland/metabolism , Animals , Cell Line , Cell Survival/physiology , Chromones/pharmacology , Insulin/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Iodine Radioisotopes , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , Symporters/analysis , Symporters/antagonists & inhibitors , Symporters/physiology , TOR Serine-Threonine Kinases , Thyroid Gland/chemistry , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyrotropin/pharmacology
18.
Bol Asoc Med P R ; 102(4): 17-24, 2010.
Article in English | MEDLINE | ID: mdl-21766543

ABSTRACT

Previous studies suggest the presence of a minicolumnopathy in autism. Minicolumnar abnormalities as well as certain migratory and proliferative defects, common to autism, may be rooted in the general mechanics of periventricular germinal cell division and maturation. Increased numbers of periventricular germinal cell/radial glia can be mimicked by a variety of different transgenic mouse models and environmental factors. These murine models and environmental factors illustrate how a fairly homogenous neuroanatomical phenotype can diverge at the genetic level. By first defining the lowest common denominator (i.e., the minicolumn) and then examining which pathways are vulnerable to involved genetic and environmental factors, we may gain a greater understanding of the pathophysiologic mechanisms underlying Autism Spectrum Conditions.


Subject(s)
Autistic Disorder/classification , Neocortex/pathology , Animals , Autistic Disorder/epidemiology , Autistic Disorder/genetics , Autistic Disorder/pathology , Cell Division , Cell Lineage , Cell Movement , Disease Models, Animal , Environmental Exposure , Epigenesis, Genetic , Genetic Heterogeneity , Hormones/adverse effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Metabolic Networks and Pathways , Mice , Mice, Transgenic , Mutation , Neocortex/drug effects , Neocortex/embryology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neuroglia/pathology , Pluripotent Stem Cells/pathology , Species Specificity , Teratogens/toxicity
19.
J Biol Chem ; 284(23): 15857-66, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19332546

ABSTRACT

During the formation of synapses, specific regions of pre- and postsynaptic cells associate to form a single functional transmission unit. In this process, synaptogenic factors are necessary to modulate pre- and postsynaptic differentiation. In mammals, different Wnt ligands operate through canonical and non-canonical Wnt pathways, and their precise functions to coordinate synapse structure and function in the mature central nervous system are still largely unknown. Here, we studied the effect of different Wnt ligands on postsynaptic organization. We found that Wnt-5a induces short term changes in the clustering of PSD-95, without affecting its total levels. Wnt-5a promotes the recruitment of PSD-95 from a diffuse dendritic cytoplasmic pool to form new PSD-95 clusters in dendritic spines. Moreover, Wnt-5a acting as a non-canonical ligand regulates PSD-95 distribution through a JNK-dependent signaling pathway, as demonstrated by using the TAT-TI-JIP peptide in mature hippocampal neurons. Finally, using adult rat hippocampal slices, we found that Wnt-5a modulates glutamatergic synaptic transmission through a postsynaptic mechanism. Our studies indicate that the Wnt-5a/JNK pathway modulates the postsynaptic region of mammalian synapse directing the clustering and distribution of the physiologically relevant scaffold protein, PSD-95.


Subject(s)
Hippocampus/physiology , Intracellular Signaling Peptides and Proteins/physiology , MAP Kinase Kinase 4/physiology , Membrane Proteins/physiology , Neurons/physiology , Wnt Proteins/physiology , Animals , Cell Line , Disks Large Homolog 4 Protein , Electrophysiology , Embryo, Mammalian , Humans , Kidney/embryology , Rats , Rats, Sprague-Dawley , Signal Transduction , Synaptic Potentials/physiology , Wnt-5a Protein
20.
Neurodegener Dis ; 5(3-4): 149-52, 2008.
Article in English | MEDLINE | ID: mdl-18322375

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most prevalent neurodegenerative disease in the growing population of elderly people. Synaptic dysfunction is an early manifestation of AD. The cellular mechanism by which beta-amyloid peptide (Abeta) affects synapses remains unclear. Abeta oligomers target synapses in cultured rat hippocampal neurons suggesting that they play a key role in the regulation of synapses. OBJECTIVE: The aim of this work is to study the effect of Abeta oligomers on the central synapses and the possible role of the Wnt signaling pathway in preventing the Abeta effects. METHODS: We used rat hippocampal neurons, immunofluorescence and western blot procedures to detect synaptic proteins. RESULTS: Abeta oligomers induced a reduction of the postsynaptic density protein 95 (PSD-95) and the NMDA glutamate receptors. We found that Wnt-5a, a noncanonical Wnt ligand, prevents the decrease triggered by Abeta oligomers in the glutamate receptor and PSD-95. CONCLUSION: Altogether, our results suggest that Abeta oligomers decrease the synaptic responses by affecting the postsynaptic region at different levels. The Wnt signaling activation prevents synaptic damage induced by Abeta, which raises the possibility of a new therapeutic intervention for the treatment of synaptic changes observed in AD.


Subject(s)
Amyloid beta-Peptides/physiology , Signal Transduction/physiology , Synapses/pathology , Wnt Proteins/physiology , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , Hippocampus/chemistry , Hippocampus/metabolism , Hippocampus/pathology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/physiology , Neurons/chemistry , Neurons/metabolism , Neurons/pathology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/physiology , Structure-Activity Relationship , Synapses/chemistry , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL