Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1109528, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36875482

RESUMO

The principal hormonal product of the thyroid gland, L-thyroxine (T4), is a prohormone for 3,3',5-triiodo-L-thyronine, T3, the major ligand of nuclear thyroid hormone receptors (TRs). At a cell surface thyroid hormone analogue receptor on cancer cell and endothelial cell plasma membrane integrin αvß3, however, T4 at physiological concentrations is biologically active and is the major ligand. At this site in solid tumor cells, T4 nongenomically initiates cell proliferation, is anti-apoptotic by multiple mechanisms, supports radioresistance and enhances cancer-related angiogenesis. In contrast, hypothyroidism has been reported clinically to slow tumor growth. At physiological levels, T3 is not biologically active at the integrin and maintenance of euthyroidism with T3 in cancer patients may be associated with slowed tumor proliferation. Against this background, we raise the possibility that host serum T4 levels that are spontaneously in the upper tertile or quartile of the normal range in cancer patients may be a factor that contributes to aggressive tumor behavior. Recent observations on tumor metastasis and tumor-associated propensity for thrombosis due to T4 also justify clinical statistical analysis for a relationship to upper tertile hormone levels. That reverse T3 (rT3) may stimulate tumor growth has recently been reported and thus the utility of adding this measurement to thyroid function testing in cancer patients requires assessment. In summary, T4 at physiological concentrations promotes tumor cell division and aggressiveness and euthyroid hypothyroxinemia arrests clinically advanced solid tumors. These findings support the clinical possibility that T4 levels in the upper tertile of the normal range require examination as a tumor supporting factor.


Assuntos
Segunda Neoplasia Primária , Neoplasias , Humanos , Glândula Tireoide , Ligantes , Tiroxina , Receptores dos Hormônios Tireóideos
2.
Metabolites ; 12(4)2022 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-35448512

RESUMO

Chemically modified forms of tetraiodothyroacetic acid (tetrac), an L-thyroxine derivative, have been shown to exert their anticancer activity at plasma membrane integrin αvß3 of tumor cells. Via a specific hormone receptor on the integrin, tetrac-based therapeutic agents modulate expression of genes relevant to cancer cell proliferation, survival and energy metabolism. P-bi-TAT, a novel bivalent tetrac-containing synthetic compound has anticancer activity in vitro and in vivo against glioblastoma multiforme (GBM) and other types of human cancers. In the current study, microarray analysis was carried out on a primary culture of human GBM cells exposed to P-bi-TAT (10-6 tetrac equivalent) for 24 h. P-bi-TAT significantly affected expression of a large panel of genes implicated in cancer cell stemness, growth, survival and angiogenesis. Recent interest elsewhere in ATP synthase as a target in GBM cells caused us to focus attention on expression of genes involved in energy metabolism. Significantly downregulated transcripts included multiple energy-metabolism-related genes: electron transport chain genes ATP5A1 (ATP synthase 1), ATP51, ATP5G2, COX6B1 (cytochrome c oxidase subunit 6B1), NDUFA8 (NADH dehydrogenase (ubiquinone) FA8), NDUFV2I and other NDUF genes. The NDUF and ATP genes are also relevant to control of oxidative phosphorylation and transcription. Qualitatively similar actions of P-bi-TAT on expression of subsets of energy-metabolism-linked genes were also detected in established human GBM and pancreatic cancer cell lines. In conclusion, acting at αvß3 integrin, P-bi-TAT caused downregulation in human cancer cells of expression of a large number of genes involved in electron transport and oxidative phosphorylation. These observations suggest that cell surface thyroid hormone receptors on αvß3 regulate expression of genes relevant to tumor cell stemness and energy metabolism.

3.
Front Cell Dev Biol ; 10: 829788, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237605

RESUMO

Doxycycline, an antibiotic, displays the inhibition of different signal transduction pathways, such as anti-inflammation and anti-proliferation, in different types of cancers. However, the anti-cancer mechanisms of doxycycline via integrin αvß3 are incompletely understood. Integrin αvß3 is a cell-surface anchor protein. It is the target for estrogen, androgen, and thyroid hormone and plays a pivotal role in the proliferation, migration, and angiogenic process in cancer cells. In our previous study, thyroxine hormones can interact with integrin αvß3 to activate the extracellular signal-regulated kinase 1/2 (ERK1/2), and upregulate programmed death-ligand 1 (PD-L1) expression. In the current study, we investigated the inhibitory effects of doxycycline on proliferation in two breast cancer cell lines, MCF-7 and MDA-MB-231 cells. Doxycycline induces concentration-dependent anti-proliferation in both breast cancer cell lines. It regulates gene expressions involved in proliferation, pro-apoptosis, and angiogenesis. Doxycycline suppresses cell cyclin D1 (CCND1) and c-Myc which play crucial roles in proliferation. It also inhibits PD-L1 gene expression. Our findings show that modulation on integrin αvß3 binding activities changed both thyroxine- and doxycycline-induced signal transductions by an integrin αvß3 inhibitor (HSDVHK-NH2). Doxycycline activates phosphorylation of focal adhesion kinase (FAK), a downstream of integrin, but inhibits the ERK1/2 phosphorylation. Regardless, doxycycline-induced FAK phosphorylation is blocked by HSDVHK-NH2. In addition, the specific mechanism of action associated with pERK1/2 inhibition via integrin αvß3 is unknown for doxycycline treatment. On the other hand, our findings indicated that inhibiting ERK1/2 activation leads to suppression of PD-L1 expression by doxycycline treatment. Furthermore, doxycycline-induced gene expressions are disturbed by a specific integrin αvß3 inhibitor (HSDVHK-NH2) or a mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinases (ERK) kinase (MAPK/ERK, MEK) inhibitor (PD98059). The results imply that doxycycline may interact with integrin αvß3 and inhibits ERK1/2 activation, thereby regulating cell proliferation and downregulating PD-L1 gene expression in estrogen receptor (ER)-negative breast cancer MDA-MB-231 cells.

4.
Endocr Res ; 47(1): 39-44, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34775877

RESUMO

BACKGROUND: Integrin αvß3 is a cell membrane structural protein whose extracellular domain contains a receptor for L-thyroxine (T4). The integrin is expressed in rapidly dividing cells and its internalization is prompted by T4. The protein binds viruses and we have raised the possibility elsewhere that action of free T4 (FT4)-when he latter is increased in the nonthyroidal illness syndrome (NTIS) known to complicate COVID-19 infecction-may enhance cellular uptke of SARS-CoV-2 and its receptor. OBJECTIVE: Because T4 also acts nongenomically via the integrin to promote platelet aggregation and angiogenesis, we suggest here that T4 may contribute to the coagulopathy and endothelial abnormalities that can develop in COVID-19 infections, particularly when the lung is primary affected. DISCUSSION AND CONCLUSIONS: Elevated FT4 has been described in the NTIS of COVID-19 patients and may be associated with increased illness severity, but the finding of FT4 elevation is inconsistent in the NTIS literature. Circulating 3,5',3'-triiodo-L-thyronine (reverse T3, rT3) are frequently elevated in NTIS. Thought to be biologically inactive, rT3in fact stimulates cancer cell proliferation via avb3 and also may increase actin polymerization. We propose here that rT3 in the NTIS complicating systemic COVIF-19 infection may support coagulation and disordered blood vessel formation via actin polymerization.


Assuntos
COVID-19 , Humanos , Integrina alfaVbeta3 , Masculino , SARS-CoV-2 , Hormônios Tireóideos , Tiroxina , Tri-Iodotironina
5.
Front Endocrinol (Lausanne) ; 13: 1109555, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36714596

RESUMO

Thyroid hormone as L-thyroxine (T4) acts nongenomically at physiological concentrations at its cancer cell surface receptor on integrin αvß3 ('thyrointegrin') to cause cancer cell proliferation. In the case of estrogen receptor (ERα)-positive breast cancer cells, T4 via the integrin promotes ERα-dependent cancer growth in the absence of estrogen. Thus, tumor growth in the post-menopausal patient with ERα-positive cancer may again be ER-dependent because of T4. Additional mechanisms by which T4 may contribute uniquely to aggressive breast cancer behavior-independently of ER-are stimulation of immune checkpoint inhibitor gene expression and of several anti-apoptosis mechanisms. These observations may call for consideration of elimination of host T4 production in breast cancer patients whose response is suboptimal to standard chemotherapy regimens. Euthyroidism in such a setting may be maintained with exogenous 3,3',5-triiodo-L-thyronine (T3).


Assuntos
Neoplasias da Mama , Glândula Tireoide , Humanos , Feminino , Glândula Tireoide/metabolismo , Neoplasias da Mama/tratamento farmacológico , Receptor alfa de Estrogênio/metabolismo , Hormônios Tireóideos/metabolismo , Tiroxina/farmacologia
6.
Endocr Relat Cancer ; 28(11): 705-713, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34432646

RESUMO

Research on the association between thyroid hormone levels and cancer mortality remains limited and inconclusive. We determined the relation of thyroid stimulating hormone (TSH), free T4 (FT4), and free T3 (FT3) levels with mortality in overall cancer and specific tumor types. Thyroid hormone levels 1-5 years prior to cancer diagnosis, as well as multiple clinical and demographic parameters, were retrospectively collected for 10,325 Israeli cancer patients, diagnosed between 2000 and 2016. Patients treated with thyroid altering medications were excluded. Cancer diagnosis was determined via the Israel National Cancer Registry. Multivariate-adjusted Cox proportional hazards model was used to assess the hazard ratios (HRs) based on thyroid hormone function for cancer mortality. A total of 5265 patients died during the follow-up period (median of 4.4 years). TSH, FT4, and FT3 levels in the hypothyroid range were associated with increase in overall mortality (adjusted HR 1.20, 1.74, 1.87, respectively). We further analyzed the association between TSH and mortality in 14 cancer subgroups. Specifically, TSH in both the hyperthyroid and hypothyroid range was associated with melanoma mortality (adjusted HR 2.20, 4.47, respectively). In conclusion, pre-diagnosis of thyroid dysfunction is associated with increased cancer mortality, a relation likely driven by specific cancer types. These findings suggest that thyroid hormones may potentially serve as prognostic markers in cancer.


Assuntos
Hipotireoidismo , Neoplasias , Doenças da Glândula Tireoide , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Estudos Retrospectivos , Doenças da Glândula Tireoide/tratamento farmacológico , Hormônios Tireóideos , Tireotropina , Tiroxina/uso terapêutico , Tri-Iodotironina
7.
J Endocr Soc ; 5(8): bvab100, 2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34195529

RESUMO

CONTEXT: Immune checkpoint inhibitors (ICIs) have gained a revolutionary role in management of many advanced malignancies. However, immune-related endocrine events (irEEs), have been associated with their use. irEEs have nonspecific clinical presentations and variable timelines, making their early diagnosis challenging. OBJECTIVE: To identify risk factors, timelines, and prognosis associated with irEEs development. DESIGN AND SETTING: Retrospective observational study within the Cleveland Clinic center. PATIENTS: Metastatic cancer adult patients who received ICIs were included. METHODS: 570 charts were reviewed to obtain information on demographics, ICIs used, endocrine toxicities, cancer response to treatment with ICI, and overall survival. MAIN OUTCOME MEASURES: Incidence of irEEs, time to irEEs development and overall survival of patients who develop irEEs. RESULTS: The final cohort included 551 patients. The median time for the diagnosis of irEEs was 9 weeks. Melanoma was associated with the highest risk for irEEs (31.3%). Ipilimumab appeared to have the highest percentage of irEEs (29.4%), including the highest risk of pituitary insufficiency (11.7%), the most severe (Grade 4 in 60%) and irreversible (100%) forms of irEEs. Forty-five percent of patients with irEEs had adequate cancer response to ICI compared to 28.3% of patients without irEEs (P = 0.002). Patients with irEEs had significantly better survival compared to patients without irEEs (P < 0.001). CONCLUSIONS: In the adult population with metastatic cancer receiving treatment with ICI, irEEs development may predict tumor response to immunotherapy and a favorable prognosis. Ipilimumab use, combination ICI therapy, and melanoma are associated with a higher incidence of irEEs.

8.
Front Endocrinol (Lausanne) ; 12: 691736, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34234745

RESUMO

L-Thyroxine (T4) is the principal ligand of the thyroid hormone analogue receptor on the extracellular domain of integrin αvß3. The integrin is overexpressed and activated in cancer cells, rapidly dividing endothelial cells, and platelets. The biologic result is that T4 at physiological concentration and without conversion to 3,3',5-triiodo-L-thyronine (T3) may stimulate cancer cell proliferation and cancer-relevant angiogenesis and platelet coagulation. Pro-thrombotic activity of T4 on platelets is postulated to support cancer-linked blood clotting and to contribute to tumor cell metastasis. We examine some of these findings as they may relate to cancers of the thyroid. Differentiated thyroid cancer cells respond to physiological levels of T4 with increased proliferation. Thus, the possibility exists that in patients with differentiated thyroid carcinomas in whom T4 administration and consequent endogenous thyrotropin suppression have failed to arrest the disease, T4 treatment may be stimulating tumor cell proliferation. In vitro studies have shown that tetraiodothyroacetic acid (tetrac), a derivative of T4, acts via the integrin to block T4 support of thyroid cancer and other solid tumor cells. Actions of T4 and tetrac or chemically modified tetrac modulate gene expression in thyroid cancer cells. T4 induces radioresistance via induction of a conformational change in the integrin in various cancer cells, although not yet established in thyroid cancer cells. The thyroid hormone receptor on integrin αvß3 mediates a number of actions of T4 on differentiated thyroid cancer cells that support the biology of the cancer. Additional studies are required to determine whether T4 acts on thyroid cancer cells.


Assuntos
Hormônios Tireóideos/metabolismo , Trifosfato de Adenosina/metabolismo , Antígeno B7-H1/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina alfaVbeta3/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia
9.
J Clin Med ; 10(8)2021 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921634

RESUMO

BACKGROUND: Chronic lymphocytic leukemia (CLL) is the most common adult leukemia. The thyroid hormones, T3 and T4, bind the αvß3 integrin and activate phosphorylates ERK (pERK). These tumor-promoting actions were reported in a number of malignancies, but not in CLL. METHODS: Primary cells from 22 CLL patients were verified for disease markers (CD5/CD19/CD23) and analyzed for αvß3 by flow cytometry (FC), ImageStream, Western blots (WB), and immunohistochemistry (IHC) in archival bone marrow (BM, n = 6) and lymph node (LN, n = 5) tissues. Selected samples (n = 8) were incubated with T3 (1-100 nM) or T4 (0.1-10 µM) for 30 min, and the expression levels of αvß3, pERK and PCNA (cell proliferation marker) were determined (WB). RESULTS: αvß3 was detected on the membrane of circulating CLL cells and in the BM but not in the LN. T3 and T4 enhanced αvß3 protein levels in primary CLL cells. Similarly, pERK and PCNA were rapidly induced in response to T3 and T4 exposure. CONCLUSIONS: αvß3 integrin is expressed on primary CLL cells and is induced by thyroid hormones. We further suggest that the hormones are mitogenic in these cells, presumably via αvß3-mediated signaling.

10.
Eur J Endocrinol ; 184(3): 477-486, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33444229

RESUMO

OBJECTIVE: The association between dysregulated thyroid hormone function and cancer risk is inconclusive, especially among different age groups and uncommon malignancies. We sought to determine the relation of TSH and free T4 levels with overall cancer risk as well as risk of specific cancer types. DESIGN AND METHODS: Data on thyroid hormone profile was collected from 375 635 Israeli patients with no prior history of cancer. Cancer cases were identified via the Israel National Cancer Registry. Cox proportional hazards model was used to assess hazard ratios for overall cancer as well as 20 cancer subgroups. RESULTS: In this study, 23 808 cases of cancer were detected over median follow up of 10.9 years. Among patients younger than 50 at inclusion, TSH in the hyperthyroid range, elevated free T4 and subclinical hyperthyroidism were associated with increased cancer risk (HR: 1.3, 1.28 and 1.31, respectively). In contrast, patients 50 or older with clinical hyperthyroidism were at lower cancer risk (HR: 0.64). Elevated TSH was associated with decreased risk of prostate cancer (HR: 0.67). Log-TSH elevation was associated with decreased risk of thyroid cancer (HR: 0.82) and increased risk of melanoma (HR: 1.11) and uterine cancer (HR: 1.27). Elevated free T4 was associated with increased lung cancer risk (HR: 1.54), while free T4 levels above the normal range and clinical hyperthyroidism were related to lower colorectal cancer risk (HR: 0.59 and 0.08, respectively). CONCLUSIONS: Thyroid hormones display opposing effects on cancer risk, based on patient age and cancer type.


Assuntos
Hormônios Tireóideos/sangue , Adulto , Feminino , Humanos , Hipertireoidismo/sangue , Hipertireoidismo/epidemiologia , Israel/epidemiologia , Masculino , Melanoma/sangue , Melanoma/epidemiologia , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Neoplasias da Próstata/sangue , Neoplasias da Próstata/epidemiologia , Tireotropina/sangue , Neoplasias Uterinas/sangue , Neoplasias Uterinas/epidemiologia
11.
Genes (Basel) ; 11(7)2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32645835

RESUMO

The clinical behavior of thyroid cancers is seen to reflect inherent transcriptional activities of mutated genes and trophic effects on tumors of circulating pituitary thyrotropin (TSH). The thyroid hormone, L-thyroxine (T4), has been shown to stimulate proliferation of a large number of different forms of cancer. This activity of T4 is mediated by a cell surface receptor on the extracellular domain of integrin αvß3. In this brief review, we describe what is known about T4 as a circulating trophic factor for differentiated (papillary and follicular) thyroid cancers. Given T4's cancer-stimulating activity in differentiated thyroid cancers, it was not surprising to find that genomic actions of T4 were anti-apoptotic. Transduction of the T4-generated signal at the integrin primarily involved mitogen-activated protein kinase (MAPK). In thyroid C cell-origin medullary carcinoma of the thyroid (MTC), effects of thyroid hormone analogues, such as tetraiodothyroacetic acid (tetrac), include pro-angiogenic and apoptosis-linked genes. Tetrac is an inhibitor of the actions of T4 at αvß3, and it is assumed, but not yet proved, that the anti-angiogenic and pro-apoptotic actions of tetrac in MTC cells are matched by T4 effects that are pro-angiogenic and anti-apoptotic. We also note that papillary thyroid carcinoma cells may express the leptin receptor, and circulating leptin from adipocytes may stimulate tumor cell proliferation. Transcription was stimulated by leptin in anaplastic, papillary, and follicular carcinomas of genes involved in invasion, such as matrix metalloproteinases (MMPs). In summary, thyroid hormone analogues may act at their receptor on integrin αvß3 in a variety of types of thyroid cancer to modulate transcription of genes relevant to tumor invasiveness, apoptosis, and angiogenesis. These effects are independent of TSH.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias da Glândula Tireoide/genética , Tiroxina/análogos & derivados , Animais , Humanos , Integrina alfaVbeta3/metabolismo , Leptina/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Tiroxina/farmacologia
12.
Endocr Res ; 45(3): 210-215, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32628899

RESUMO

BACKGROUND: Uptake of coronaviruses by target cells involves binding of the virus by cell ectoenzymes. For the etiologic agent of COVID-19 (SARS-CoV-2), a receptor has been identified as angiotensin-converting enzyme-2 (ACE2). Recently it has been suggested that plasma membrane integrins may be involved in the internalization and replication of clinically important coronaviruses. For example, integrin αvß3 is involved in the cell uptake of a model porcine enteric α-coronavirus that causes human epidemics. ACE2 modulates the intracellular signaling generated by integrins. OBJECTIVE: We propose that the cellular internalization of αvß3 applies to uptake of coronaviruses bound to the integrin, and we evaluate the possibility that clinical host T4 may contribute to target cell uptake of coronavirus and to the consequence of cell uptake of the virus. DISCUSSION AND CONCLUSIONS: The viral binding domain of the integrin is near the Arg-Gly-Asp (RGD) peptide-binding site and RGD molecules can affect virus binding. In this same locale on integrin αvß3 is the receptor for thyroid hormone analogues, particularly, L-thyroxine (T4). By binding to the integrin, T4 has been shown to modulate the affinity of the integrin for other proteins, to control internalization of αvß3 and to regulate the expression of a panel of cytokine genes, some of which are components of the 'cytokine storm' of viral infections. If T4 does influence coronavirus uptake by target cells, other thyroid hormone analogues, such as deaminated T4 and deaminated 3,5,3'-triiodo-L-thyronine (T3), are candidate agents to block the virus-relevant actions of T4 at integrin αvß3 and possibly restrict virus uptake.


Assuntos
Infecções por Coronavirus/virologia , Integrina alfaVbeta3/metabolismo , Vírus da Diarreia Epidêmica Suína/metabolismo , Receptores Virais/efeitos dos fármacos , Hormônios Tireóideos/farmacologia , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/metabolismo , Sítios de Ligação , COVID-19 , Citocinas/fisiologia , Células Epiteliais/virologia , Humanos , Oligopeptídeos/metabolismo , Pandemias , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/virologia , Receptores Virais/química , Receptores Virais/metabolismo , SARS-CoV-2 , Suínos , Hormônios Tireóideos/fisiologia , Tiroxina/fisiologia , Internalização do Vírus
13.
Artigo em Inglês | MEDLINE | ID: mdl-31507530

RESUMO

Hypothyroidism has been reported to improve survival in cancer patients but only recently has the putative mechanism been identified as a receptor for thyroxine and tri-iodothyronine on integrin αvß3. Recognition of divergence of action of the pro-oncogenic L-thyroxine (T4) from pro-metabolic 3,5,3'-triiodo-L-thyronine (T3) has enabled clinical implementation whereby exogenous T3 may replace exogenous (or endogenous) T4 to maintain clinical euthyroid hypothyroxinemia that results in significantly better survival in advanced cancer patients without the morbidity of clinical hypothyroidism.

14.
Endocr Res ; 44(4): 148-152, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30943372

RESUMO

Background: Reverse T3 (rT3; 3,3',5'-triiodo-L-thyronine) is widely regarded as an inactive naturally occurring analog of thyroid hormone. rT3 is known to bind to the thyroid hormone analog receptor on plasma membrane integrin αvß3. This integrin is generously expressed by tumor cells and is the initiation site for the stimulation by L-thyroxine (T4) at physiological free concentrations on cancer cell proliferation. Results: In the present studies, we show that rT3 caused increases of proliferation in vitro of 50% to 80% (P < 0.05-0.001) of human breast cancer and glioblastoma cells. Conclusion: rT3 may be a host factor supporting cancer growth.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias/patologia , Tri-Iodotironina Reversa/farmacologia , Adenocarcinoma/patologia , Neoplasias Encefálicas/patologia , Neoplasias da Mama/patologia , Relação Dose-Resposta a Droga , Feminino , Glioblastoma/patologia , Humanos , Células MCF-7 , Células Tumorais Cultivadas
15.
Discov Med ; 27(147): 111-117, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30939295

RESUMO

The classical molecular mechanism of thyroid hormone involves the intranuclear interaction of 3,5,3'-triiodo-L-thyronine (T3) with thyroid hormone-specific nuclear proteins and consequent specific gene expression. This mechanism prevails in normal cells. What we emphasize here is that how thyroid hormone acts depends upon the types of cell or cell-like structure, e.g., platelet, under consideration, and that cancer cells, dividing endothelial cells, phagocytes, and platelets respond to the liganding of L-thyroxine (T4) by plasma membrane integrin αvß3. In intact tumor cells, T4 at the integrin can modulate the transcription of a substantial number of specific genes relevant to cancer cell proliferation, cell metabolism, and cancer cell anti-apoptosis defense. T4 may also regulate the interactions of the integrin in the endothelial cell plasma membrane with adjacent vascular growth factor receptors, modulating angiogenesis. T4 activates platelets via αvß3 transferred from the megakaryocyte. It is also possible that, in addition to T4, reverse T3 (rT3) may have actions in cancer cells at the thyroid hormone receptor on αvß3.


Assuntos
Membrana Celular/metabolismo , Proliferação de Células , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Tiroxina/metabolismo , Animais , Membrana Celular/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina alfaVbeta3/metabolismo , Neoplasias/patologia
16.
Artigo em Inglês | MEDLINE | ID: mdl-30564196

RESUMO

In the context of genomic thyroid hormone actions in normal (noncancer) cells that involve primary interactions with nuclear thyroid hormone receptors (TRs), L-thyroxine (T4), and 3,3',5'-triiodo-L-thyronine (reverse T3, rT3) have little bioactivity. In terms of TRs, T4 is a prohormone from which the active nuclear ligand, 3,5,3'-triido-L-thyronine (T3), is generated by deiodination. Deaminated T4 and T3 metabolites have different genomic effects: tetraiodothyroacetic acid (tetrac) is a low grade thyromimetic derivative of T4, whereas triiodothyroacetic acid (triac), the acetic acid metabolite of T3, has substantial thyromimetic activity. In cancer cells, the cell surface receptor for thyroid hormone on integrin αvß3 mediates non-genomic actions of thyroid hormone analogs. The integrin is expressed in large measure by cancer cells and dividing endothelial cells and has a substantially different panel of responses to thyroid hormone analogs. At αvß3, T4 is a potent proliferative, anti-apoptotic and pro-angiogenic hormone and is the primary ligand. rT3 may also be proliferative at this site. In contrast, tetrac and triac are antagonists of T4 at αvß3, but also have anticancer properties at this site that are independent of their effects on the binding of T4.

17.
Biomedicines ; 6(3)2018 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-30135398

RESUMO

Acting at a cell surface receptor on the extracellular domain of integrin αvß3, thyroid hormone analogues regulate downstream the expression of a large panel of genes relevant to cancer cell proliferation, to cancer cell survival pathways, and to tumor-linked angiogenesis. Because αvß3 is involved in the cancer cell metastatic process, we examine here the possibility that thyroid hormone as l-thyroxine (T4) and the thyroid hormone antagonist, tetraiodothyroacetic acid (tetrac), may respectively promote and inhibit metastasis. Actions of T4 and tetrac that are relevant to cancer metastasis include the multitude of synergistic effects on molecular levels such as expression of matrix metalloproteinase genes, angiogenesis support genes, receptor tyrosine kinase (EGFR/ERBB2) genes, specific microRNAs, the epithelial⁻mesenchymal transition (EMT) process; and on the cellular level are exemplified by effects on macrophages. We conclude that the thyroid hormone-αvß3 interaction is mechanistically linked to cancer metastasis and that modified tetrac molecules have antimetastatic activity with feasible therapeutic potential.

18.
Horm Cancer ; 9(5): 349-360, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30027502

RESUMO

Cancer resistance to chemotherapeutic agents is a major issue in the management of cancer patients. Overexpression of the ribonucleotide reductase regulatory subunit M2 (RRM2) has been associated with aggressive cancer behavior and chemoresistance. Nano-diamino-tetrac (NDAT) is a nanoparticulate derivative of tetraiodothyroacetic acid (tetrac), which exerts anticancer properties via several mechanisms and downregulates RRM2 gene expression in cancer cells. Resveratrol is a stilbenoid phytoalexin which binds to a specific site on the cell surface integrin αvß3 to trigger cancer cell death via nuclear translocation of COX-2. Here we report that resveratrol paradoxically activates RRM2 gene expression and protein translation in colon cancer cells. This unanticipated effect inhibits resveratrol-induced COX-2 nuclear accumulation. RRM2 downregulation, whether achieved by RNA interference or treatment with NDAT, enhanced resveratrol-induced COX-2 gene expression and nuclear uptake which is essential to integrin αvß3-mediated-resveratrol-induced antiproliferation in cancer cells. Elsewhere, NDAT downregulated resveratrol-induced RRM2 expression in vivo but potentiated the anticancer effect of the stilbene. These findings suggest that RRM2 appears as a cancer cell defense mechanism which can hinder the anticancer effect of the stilbene via the integrin αvß3 axis. Furthermore, the antagonistic effect of RRM2 against resveratrol is counteracted by the administration of NDAT.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Neoplasias Colorretais/genética , Poliglactina 910/uso terapêutico , Resveratrol/uso terapêutico , Tiroxina/análogos & derivados , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Nus , Poliglactina 910/farmacologia , Resveratrol/farmacologia , Tiroxina/farmacologia , Tiroxina/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Endocr Res ; 43(4): 215-219, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29611723

RESUMO

PURPOSE: Integrin αvß3 is an important structural and signaling protein of the plasma membrane of cancer cells and dividing blood vessel cells. The plastic extracellular domain of the protein binds to extracellular matrix proteins and plasma membrane proteins, changing cell-cell interactions and generating intracellular signals that influence cell behavior. αvß3 also contains a receptor for thyroid hormone and derivatives, including tetraiodothyroacetic acid (tetrac). MATERIALS AND METHODS: Human prostate cancer (PC3) cells were engrafted in the chicken chorioallantoic membrane model. The well-vascularized spheroidal xenografts were exposed to X-radiation in varying dosages (1-10 Gy) and in the presence and absence of an antibody that recognizes unliganded human ß3 integrin monomer in the extended or open (activated) configuration. RESULTS: Radiation significantly increased activated ß3 within 1 h (P < .001), a radiation response not previously reported. Incubation of cells with unmodified tetrac or tetrac covalently linked to a nanoparticle (Nanotetrac, NDAT) did not change basal activation state of the integrin monomer, but prevented radiation-induced activation of ß3. CONCLUSIONS: Activation of the integrin in response to radiation is interpreted as a defensive response, perhaps leading to increased intercellular affinity and inhibition of cell division, a radioresistant state. Action of NDAT indicates that pharmacologic interventions in the radiation response of integrin ß3 monomer and therefore of αvß3 are feasible.


Assuntos
Membrana Corioalantoide/metabolismo , Integrina alfaVbeta3/metabolismo , Próstata/metabolismo , Tiroxina/análogos & derivados , Animais , Galinhas , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/efeitos da radiação , Humanos , Masculino , Células PC-3 , Próstata/efeitos dos fármacos , Próstata/efeitos da radiação , Tiroxina/farmacologia
20.
Endocr Relat Cancer ; 25(5): 533-545, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29555649

RESUMO

Thyroid hormone, l-thyroxine (T4), has been shown to promote ovarian cancer cell proliferation via a receptor on plasma membrane integrin αvß3 and to induce the activation of ERK1/2 and expression of programmed death-ligand 1 (PD-L1) in cancer cells. In contrast, resveratrol binds to integrin αvß3 at a discrete site and induces p53-dependent antiproliferation in malignant neoplastic cells. The mechanism of resveratrol action requires nuclear accumulation of inducible cyclooxygenase (COX)-2 and its complexation with phosphorylated ERK1/2. In this study, we examined the mechanism by which T4 impairs resveratrol-induced antiproliferation in human ovarian cancer cells and found that T4 inhibited resveratrol-induced nuclear accumulation of COX-2. Furthermore, T4 increased expression and cytoplasmic accumulation of PD-L1, which in turn acted to retain inducible COX-2 in the cytoplasm. Knockdown of PD-L1 by small hairpin RNA (shRNA) relieved the inhibitory effect of T4 on resveratrol-induced nuclear accumulation of COX-2- and COX-2/p53-dependent gene expression. Thus, T4 inhibits COX-2-dependent apoptosis in ovarian cancer cells by retaining inducible COX-2 with PD-L1 in the cytoplasm. These findings provide new insights into the antagonizing effect of T4 on resveratrol's anticancer properties.


Assuntos
Apoptose/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Resveratrol/metabolismo , Tiroxina/uso terapêutico , Feminino , Humanos , Neoplasias Ovarianas/patologia , Tiroxina/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA