Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Nutr ; 61(2): 625-636, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34687321

RESUMO

PURPOSE: In less than one and a half year, the COVID-19 pandemic has nearly brought to a collapse our health care and economic systems. The scientific research community has concentrated all possible efforts to understand the pathogenesis of this complex disease, and several groups have recently emphasized recommendations for nutritional support in COVID-19 patients. In this scoping review, we aim at encouraging a deeper appreciation of magnesium in clinical nutrition, in view of the vital role of magnesium and the numerous links between the pathophysiology of SARS-CoV-2 infection and magnesium-dependent functions. METHODS: By searching PubMed and Google Scholar from 1990 to date, we review existing evidence from experimental and clinical studies on the role of magnesium in chronic non-communicable diseases and infectious diseases, and we focus on recent reports of alterations of magnesium homeostasis in COVID-19 patients and their association with disease outcomes. Importantly, we conduct a census on ongoing clinical trials specifically dedicated to disclosing the role of magnesium in COVID-19. RESULTS: Despite many methodological limitations, existing data seem to corroborate an association between deranged magnesium homeostasis and COVID-19, and call for further and better studies to explore the prophylactic or therapeutic potential of magnesium supplementation. CONCLUSION: We propose to reconsider the relevance of magnesium, frequently overlooked in clinical practice. Therefore, magnesemia should be monitored and, in case of imbalanced magnesium homeostasis, an appropriate nutritional regimen or supplementation might contribute to protect against SARS-CoV-2 infection, reduce severity of COVID-19 symptoms and facilitate the recovery after the acute phase.


Assuntos
COVID-19 , Homeostase , Humanos , Magnésio , Pandemias , SARS-CoV-2
2.
Eur J Nutr ; 61(7): 3697-3706, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35689124

RESUMO

PURPOSE: Serum magnesium is the most frequently used laboratory test for evaluating clinical magnesium status. Hypomagnesemia (low magnesium status), which is associated with many chronic diseases, is diagnosed using the serum magnesium reference range. Currently, no international consensus for a magnesemia normal range exists. Two independent groups designated 0.85 mmol/L (2.07 mg/dL; 1.7 mEq/L) as the low cut-off point defining hypomagnesemia. MaGNet discussions revealed differences in serum magnesium reference ranges used by members' hospitals and laboratories, presenting an urgent need for standardization. METHODS: We gathered and compared serum magnesium reference range values from our institutions, hospitals, and colleagues worldwide. RESULTS: Serum magnesium levels designating "hypomagnesemia" differ widely. Of 43 collected values, only 2 met 0.85 mmol/L as the low cut-off point to define hypomagnesemia. The remainder had lower cut-off values, which may underestimate hypomagnesemia diagnosis in hospital, clinical, and research assessments. Current serum magnesium reference ranges stem from "normal" populations, which unknowingly include persons with chronic latent magnesium deficit (CLMD). Serum magnesium levels of patients with CLMD fall within widely used "normal" ranges, but their magnesium status is too low for long-term health. The lower serum magnesium reference (0.85 mmol/L) proposed specifically prevents the inclusion of patients with CLMD. CONCLUSIONS: Widely varying serum magnesium reference ranges render our use of this important medical tool imprecise, minimizing impacts of low magnesium status or hypomagnesemia as a marker of disease risk. To appropriately diagnose, increase awareness of, and manage magnesium status, it is critical to standardize lower reference values for serum magnesium at 0.85 mmol/L (2.07 mg/dL; 1.7 mEq/L).


Assuntos
Magnésio , Humanos , Padrões de Referência , Valores de Referência
4.
Med Teach ; 39(9): 981-986, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28552022

RESUMO

CONTEXT: The emerging global-health paradigm requires medical teaching to be continuously redefined and updated; to this end, transnational approaches should be encouraged and medical training harmonized. Infectious diseases (ID) teaching in the current context of emerging infections, fast-increasing bacterial resistance and large-scale human migration, was chosen to develop a common international course. OBJECTIVE: We report the successful implementation of a joint European undergraduate course aiming to (i) develop a common ID core curriculum among European medical schools; (ii) promote mobility among teachers and students (iii) promote international cooperation among European teachers. METHODS: The course was built around teachers' mobility. It was delivered in English by a team of European medical educators from Paris Descartes University, Università Cattolica del Sacro Cuore in Rome and the University of Edinburgh to groups of 25-30 undergraduate medical students at each university. Partner Institutions officially recognized the course as substitutive of or additive to the regular curriculum. RESULTS: The course has been running for 3 years and received excellent satisfaction scores by students and staff as regards to scientific content, pedagogy and international exchanges. CONCLUSION: This cooperative approach demonstrates the feasibility of a harmonized European undergraduate medical education, having ID as a test experiment for future developments.


Assuntos
Doenças Transmissíveis , Currículo , Educação de Graduação em Medicina/métodos , Saúde Global/educação , Estudantes de Medicina , Ensino , Doenças Transmissíveis Emergentes , Farmacorresistência Bacteriana , Educação Médica , Europa (Continente) , Humanos , Saúde Pública , Faculdades de Medicina , Migrantes
5.
Biochim Biophys Acta ; 1848(10 Pt B): 2727-37, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25596534

RESUMO

Antitumor drugs have long been known to introduce a measurable risk of cardiovascular events. Cardio-Oncology is the discipline that builds on collaboration between cardiologists and oncologists and aims at screening, preventing or minimizing such a risk. Overt concern about "possible" cardiovascular toxicity might expose cancer patients to the risk of tumor undertreatment and poor oncologic outcome. Careful analysis of risk:benefit balance is therefore central to the management of patients exposed to potentially cardiotoxic drugs. Concomitant or sequential management of cardiac and cancer therapies should also be tailored to the following strengths and weaknesses: i) molecular mechanisms and clinical correlates of cardiotoxicity have been characterized to some extent for anthracyclines but not for other chemotherapeutics or new generation "targeted" drugs, ii) anthracyclines and targeted drugs cause different mechanisms of cardiotoxicity (type I versus type II), and this classification should guide strategies of primary or secondary prevention, iii) with anthracyclines and nonanthracycline chemotherapeutics, cardiovascular events may occur on treatment as well as years or decades after completing chemotherapy, iv) some patients may be predisposed to a higher risk of cardiac events but there is a lack of prospective studies that characterized optimal genetic tests and pharmacologic measures to minimize excess risk, v) clinical toxicity may be preceded by asymptomatic systolic and/or diastolic dysfunction that necessitates innovative mechanism-based pharmacologic treatment, and vi) patient-tailored pharmacologic correction of comorbidities is important for both primary and secondary prevention. Active collaboration of physicians with laboratory scientists is much needed for improving management of cardiovascular sequelae of antitumor therapy. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Assuntos
Antiarrítmicos/uso terapêutico , Cardiotônicos/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Gerenciamento Clínico , Neoplasias/tratamento farmacológico , Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Cardiotoxinas/uso terapêutico , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/patologia , Esquema de Medicação , Inibidores Enzimáticos/uso terapêutico , Humanos , Neoplasias/complicações , Neoplasias/patologia , Medição de Risco
6.
Chem Res Toxicol ; 29(3): 317-22, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-26918850

RESUMO

Magnesium is directly involved in the control of cell growth and survival, but its role in cancer biology and therapy is multifaceted; in particular, it is highly controversial whether magnesium levels can affect therapy outcomes. Here we investigated whether magnesium availability can modulate cellular responses to the widely used chemotherapeutic doxorubicin. We used an in vitro model consisting of mammary epithelial HC11 cells and found that high magnesium availability was correlated with diminished sensitivity both in cells chronically adapted to high magnesium concentrations and in acutely magnesium-supplemented cells. This decrease in sensitivity resulted from reduced intracellular doxorubicin accumulation in the face of a similar drug uptake rate. We observed that high-magnesium conditions caused a decrease in intracellular drug retention by altering drug lysosomal sequestration and trafficking. In our model, magnesium supplementation correspondingly modulated expression of the TRPM7 channel, which is known to control cytoskeletal organization and dynamics and may be involved in the proposed mechanism. Our findings suggest that magnesium supplementation in hypomagnesemic cancer patients may hinder response to therapy.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Magnésio/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Células MCF-7 , Proteínas Serina-Treonina Quinases/metabolismo , Relação Estrutura-Atividade , Canais de Cátion TRPM/metabolismo
7.
Hum Mutat ; 35(1): 58-62, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24130152

RESUMO

A 0.8 kb intronic duplication in MAGT1 and a single base pair deletion in the last exon of ATRX were identified using a chromosome X-specific microarray and exome sequencing in a family with five males demonstrating intellectual disability (ID) and unusual skin findings (e.g., generalized pruritus). MAGT1 is an Mg²âº transporter previously associated with primary immunodeficiency and ID, whereas mutations in ATRX cause ATRX-ID syndrome. In patient cells, the function of ATRX was demonstrated to be abnormal based on altered RNA/protein expression, hypomethylation of rDNA, and abnormal cytokinesis. Dysfunction of MAGT1 was reflected in reduced RNA/protein expression and Mg²âº influx. The mutation in ATRX most likely explains the ID, whereas MAGT1 disruption could be linked to abnormal skin findings, as normal magnesium homeostasis is necessary for skin health. This work supports observations that multiple mutations collectively contribute to the phenotypic variability of syndromic ID, and emphasizes the importance of correlating clinical phenotype with genomic and cell function analyses.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Prurido/genética , Cromossomos Humanos X , Citocinese , Metilação de DNA , DNA Ribossômico/metabolismo , Exoma , Feminino , Genes Duplicados , Humanos , Íntrons , Magnésio/metabolismo , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , Fenótipo , Mutação Puntual , Prurido/patologia , Análise de Sequência de DNA , Síndrome , Proteína Nuclear Ligada ao X
8.
Biochem Biophys Res Commun ; 454(4): 572-5, 2014 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-25450695

RESUMO

Magnesium is well established as a fundamental factor that regulates cell proliferation. However, the molecular mechanisms linking mitogenic signals, extracellular magnesium availability and intracellular effectors are still largely unknown. In the present study we sought to determine whether EGF regulates magnesium homeostasis in normal HC11 mammary epithelial cells. To this end, we measured Mg(2+) and Ca(2+) fluxes by confocal imaging in live cells loaded with specific fluorescent ion indicators (Mag-Fluo-4 and Fluo-4, respectively). EGF stimulation induces a rapid and sustained increase in intracellular Mg(2+), concomitantly with a rise in intracellular calcium. The increase in intracellular Mg(2+) derives from an influx from the extracellular compartment, and does not depend on Ca(2+). On the contrary, the increase in intracellular Ca(2+) derives from intracellular stores, and is impaired in the absence of extracellular magnesium. Inhibition of the EGF receptor tyrosine kinase by Tyrphostin AG1478 markedly inhibits EGF-induced Mg(2+) and Ca(2+) signals. These findings demonstrate that not only does Mg(2+) influx represent an important step in the physiological response of epithelial cells to EGF, but unexpectedly the EGF-induced Mg(2+) influx is essential for the Ca(2+) signal to occur.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Magnésio/metabolismo , Animais , Células Cultivadas , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C
9.
Magnes Res ; 0(0)2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38652733

RESUMO

Since the start of the COVID-19 pandemic, it has become increasingly clear that the disease can have relevant multisystemic and long-term effects, and several studies have attempted to identify key determinants of the disease course. Here we discuss recent evidence suggesting that, in long COVID patients, combined magnesium and vitamin D deficiencies associate with a higher number of clinical manifestations, as compared to patients with normal levels of both nutrients. We highlight the potential synergistic effects of these deficiencies and propose that future studies should explore a causal link with the risk of developing long COVID. Most importantly, randomized clinical trials are needed to determine if magnesium and vitamin D supplementation can improve long COVID symptoms, providing a safe and affordable support therapy to the benefit of patients and society.

10.
Nutrients ; 16(10)2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38794644

RESUMO

Endothelial dysfunction is a crucial event in the early pathogenesis of cardiovascular diseases and is linked to magnesium (Mg) deficiency. Indeed, in endothelial cells, low Mg levels promote the acquisition of a pro-inflammatory and pro-atherogenic phenotype. This paper investigates the mechanisms by which Mg deficiency promotes oxidative stress and affects endothelial behavior in human umbilical vascular endothelial cells (HUVECs). Our data show that low Mg levels trigger oxidative stress initially by increasing NAPDH oxidase activity and then by upregulating the pro-oxidant thioredoxin-interacting protein TXNIP. The overproduction of reactive oxygen species (ROS) activates NF-κB, leading to its increased binding to the inducible nitric oxide synthase (iNOS) promoter, with the consequent increase in iNOS expression. The increased levels of nitric oxide (NO) generated by upregulated iNOS contribute to disrupting endothelial cell function by inhibiting growth and increasing permeability. In conclusion, we provide evidence that multiple mechanisms contribute to generate a pro-oxidant state under low-Mg conditions, ultimately affecting endothelial physiology. These data add support to the notion that adequate Mg levels play a significant role in preserving cardiovascular health and may suggest new approaches to prevent or manage cardiovascular diseases.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Deficiência de Magnésio , Magnésio , Óxido Nítrico Sintase Tipo II , Óxido Nítrico , Estresse Oxidativo , Espécies Reativas de Oxigênio , Humanos , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Deficiência de Magnésio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Óxido Nítrico/metabolismo , Magnésio/metabolismo , NF-kappa B/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Endotélio Vascular/metabolismo
11.
Clin Sci (Lond) ; 123(7): 417-27, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22671428

RESUMO

The relationship between magnesium and cancer is not as simple as could be assumed from the well-established requirement of magnesium for cell proliferation. Basic and pre-clinical studies indicate that magnesium deficiency can have both anti- and pro-tumour effects. In the present review, we briefly outline the new findings on the role of magnesium in angiogenesis and metastatization, and focus on the relationship between tumour cell proliferation and metabolic reprogramming, discussing how magnesium and its transporters are involved in these processes. The role of magnesium in cancer is also critically examined with regard to mitochondrial function, apoptosis and resistance to treatment. Finally, we bring together the latest experimental evidence indicating that alteration in the expression and/or activity of magnesium channels is a frequent finding in cancer cells and human tumour tissues examined to date, and we discuss the potential implications for developing novel diagnostic and therapeutic strategies.


Assuntos
Magnésio/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Canais de Cátion TRPM/metabolismo , Animais , Humanos , Proteínas Serina-Treonina Quinases
12.
Invest New Drugs ; 29(1): 73-86, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19834646

RESUMO

The cellular effects of a novel DNA-intercalating agent, the bipyridyl complex of platinum(II) with diphenyl thiourea, [Pt(bipy)(Ph(2)-tu)(2)]Cl(2), has been analyzed in the cisplatin (cDDP)-sensitive human ovarian carcinoma cell line, 2008, and its -resistant variant, C13* cells, in which the highest accumulation and cytotoxicity was found among six related bipyridyl thiourea complexes. We also show here that this complex causes reactive oxygen species to form and inhibits topoisomerase II activity to a greater extent in the sensitive than in the resistant line. The impairment of this enzyme led to DNA damage, as shown by the comet assay. As a consequence, cell cycle distribution has also been greatly perturbed in both lines. Morphological analysis revealed deep cellular derangement with the presence of cellular masses, together with increased membrane permeability and depolarization of the mitochondrial membrane. Some of these effects, sometimes differentially evident between the two cell lines, might also be related to the decrease of total cell magnesium content caused by this thiourea complex both in sensitive and resistant cells, though the basal content of this ion was higher in the cDDP-resistant line. Altogether these results suggest that this compound exerts its cytotoxicity by mechanisms partly mediated by the resistance phenotype. In particular, cDDP-sensitive cells were affected mostly by impairing topoisomerase II activity and by increasing membrane permeability and the formation of reactive oxygen species; conversely, mitochondrial impairment appeared to play the most important role in the action of complex F in resistant cells.


Assuntos
2,2'-Dipiridil/farmacologia , Cisplatino/farmacologia , DNA de Neoplasias/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Substâncias Intercalantes/farmacologia , Compostos Organoplatínicos/farmacologia , Neoplasias Ovarianas/patologia , 2,2'-Dipiridil/química , Ciclo Celular/efeitos dos fármacos , Fusão Celular , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Substâncias Intercalantes/química , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Magnésio/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Compostos Organoplatínicos/química , Neoplasias Ovarianas/enzimologia , Espécies Reativas de Oxigênio/metabolismo
13.
Nutrients ; 13(12)2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34959740

RESUMO

Nutritional deficiencies are common in inflammatory bowel diseases (IBD). In patients, magnesium (Mg) deficiency is associated with disease severity, while in murine models, dietary Mg supplementation contributes to restoring mucosal function. Since Mg availability modulates key bacterial functions, including growth and virulence, we investigated whether the beneficial effects of Mg supplementation during colitis might be mediated by gut microbiota. The effects of dietary Mg modulation were assessed in a murine model of dextran sodium sulfate (DSS)-induced colitis by monitoring magnesemia, weight, and fecal consistency. Gut microbiota were analyzed by 16S-rRNA based profiling on fecal samples. Mg supplementation improved microbiota richness in colitic mice, increased abundance of Bifidobacterium and reduced Enterobacteriaceae. KEEG pathway analysis predicted an increase in biosynthetic metabolism, DNA repair and translation pathways during Mg supplementation and in the presence of colitis, while low Mg conditions favored catabolic processes. Thus, dietary Mg supplementation increases bacteria involved in intestinal health and metabolic homeostasis, and reduces bacteria involved in inflammation and associated with human diseases, such as IBD. These findings suggest that Mg supplementation may be a safe and cost-effective strategy to ameliorate disease symptoms and restore a beneficial intestinal flora in IBD patients.


Assuntos
Colite/microbiologia , Colite/terapia , Microbioma Gastrointestinal/efeitos dos fármacos , Magnésio/farmacologia , Animais , Colite/induzido quimicamente , Sulfato de Dextrana , Modelos Animais de Doenças , Disbiose/microbiologia , Disbiose/terapia , Fezes/microbiologia , Feminino , Deficiência de Magnésio/microbiologia , Deficiência de Magnésio/terapia , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S
14.
J Cell Physiol ; 222(2): 374-81, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19890837

RESUMO

Mammary epithelial cells (HC11) chronically adapted to grow in a low-magnesium (0.05 mM vs. 0.5 mM) or in a high-magnesium (40 mM) medium were used to investigate on the mechanisms of cell magnesium transport under conditions of non-physiological magnesium availability. Magnesium influx was higher in low-magnesium cells compared to control or high-magnesium cells, whereas magnesium efflux was higher in high-magnesium cells compared to control and low-magnesium cells. Magnesium efflux was partially inhibited by imipramine, inhibitor of the Na(+)/Mg(2+) exchange. Using a monoclonal antibody detecting a approximately 70 kDa protein associated with Na(+)/Mg(2+) exchange activity, we found that the expression levels of this protein were proportional to magnesium efflux capacity, that is, high-magnesium cells > control cells > low-magnesium cells. As for magnesium influx, this was abolished by Co(III)hexaammine, inhibitor of magnesium channels. Surprisingly, we found that cells grown in low magnesium upregulated mRNA for the magnesium channel TRPM6, but not for other channels like TRPM7 or MagT1. TRPM6 mRNA was also rapidly upregulated or downregulated in HC11 cells deprived of magnesium or in low-magnesium cells re-added with magnesium, respectively. TRPM6 protein levels, as assessed by Western blot and immunofluorescence, underwent similar changes under comparable conditions. We propose that mammary epithelial cells adapt to decreased magnesium availability by upregulating magnesium influx via TRPM6, and counteract increased magnesium availability by increasing magnesium efflux primarily via Na(+)/Mg(2+) exchange. These results show, for the first time, that TRPM6 contributes to regulating magnesium influx in mammary epithelial cells, similar to what is known for intestine and kidney.


Assuntos
Antiporters/metabolismo , Células Epiteliais/metabolismo , Magnésio/metabolismo , Glândulas Mamárias Animais/metabolismo , Sódio/metabolismo , Canais de Cátion TRPM/metabolismo , Adaptação Fisiológica , Animais , Antiporters/antagonistas & inibidores , Transporte Biológico , Western Blotting , Linhagem Celular , Cobalto/farmacologia , Células Epiteliais/efeitos dos fármacos , Feminino , Imunofluorescência , Imipramina/farmacologia , Cinética , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Microscopia Confocal , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética
15.
Analyst ; 135(8): 1855-66, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20544083

RESUMO

A wide variety of biochemical reactions and physiological functions are known to require magnesium; nonetheless, its regulatory mechanisms (both at the cellular and systemic level) are still poorly characterised. Derangement of magnesium homeostasis is associated with several relevant human pathologies, e.g. diabetes, neuromuscular disorders, hypertension and other cardiovascular diseases. The study of the regulation of magnesium has gained particular interest in the last decades thanks to the molecular characterisation of specific magnesium transporters and the exploitation of molecular biology techniques to clarify their cellular and physiological function(s). In contrast, experimental tools to trace cellular magnesium and to define its homeostasis in living cells have not witnessed a corresponding progress. It was not until recently that efforts were paid to design more appropriate fluorescent indicators that could translate the advances of live imaging techniques into the field of magnesium research. Herein we critically summarise the state of the art in intracellular magnesium detection by fluorescent probes and focus on the need for improving methods and techniques in this area. We highlight the advantages of last-generation fluorescent indicators and discuss a number of challenges and opportunities that the development of novel and better sensors for magnesium still faces.


Assuntos
Espaço Intracelular/química , Magnésio/análise , Imagem Molecular/métodos , Corantes Fluorescentes/análise , Corantes Fluorescentes/química , Humanos , Espaço Intracelular/metabolismo , Magnésio/metabolismo , Espectrometria de Fluorescência
16.
Magnes Res ; 33(2): 21-27, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32554340

RESUMO

More and more studies are accumulating about COVID-19. Some aspects of the pathogenesis of the disease recall events occurring in Mg deficiency, such as a drop of T cells, increased plasma concentration of inflammatory cytokines, and endothelial dysfunction. We hypothesize that a low Mg status, which is rather common, might foment the transition from mild to critical clinical manifestations of the disease. Epidemiological, clinical, and fundamental research is needed to clarify the potential role of Mg deficiency in COVID-19.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/sangue , Deficiência de Magnésio/sangue , Magnésio/sangue , Pneumonia Viral/sangue , Animais , COVID-19 , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Progressão da Doença , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/sangue , Deficiência de Magnésio/epidemiologia , Deficiência de Magnésio/imunologia , Deficiência de Magnésio/virologia , Pandemias , Pneumonia Viral/epidemiologia , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Prognóstico , SARS-CoV-2 , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Fatores de Tempo
17.
Nutrients ; 12(11)2020 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-33114586

RESUMO

Hypomagnesemia is very commonly observed in cancer patients, most frequently in association with therapy with cetuximab (CTX), a monoclonal antibody targeting the epithelial growth factor receptor (EGFR). CTX-induced hypomagnesemia has been ascribed to renal magnesium (Mg) wasting. Here, we sought to clarify whether CTX may also influence intestinal Mg absorption and if Mg supplementation may interfere with CTX activity. We used human colon carcinoma CaCo-2 cells as an in vitro model to study the mechanisms underlying Mg transport and CTX activity. Our findings demonstrate that TRPM6 is the key channel that mediates Mg influx in intestinal cells and that EGF stimulates such influx; consequently, CTX downregulates TRPM6-mediated Mg influx by interfering with EGF signaling. Moreover, we show that Mg supplementation does not modify either the CTX IC50 or CTX-dependent inhibition of ERK1/2 phosphorylation. Our results suggest that reduced Mg absorption in the intestine may contribute to the severe hypomagnesemia that occurs in CTX-treated patients, and Mg supplementation may represent a safe and effective nutritional intervention to restore Mg status without impairing the CTX efficacy.


Assuntos
Cetuximab/efeitos adversos , Fator de Crescimento Epidérmico/metabolismo , Absorção Intestinal/efeitos dos fármacos , Magnésio/metabolismo , Canais de Cátion TRPM/metabolismo , Células CACO-2 , Colo/metabolismo , Humanos , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Erros Inatos do Transporte Tubular Renal/metabolismo
18.
Dig Liver Dis ; 52(10): 1188-1194, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32505565

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) predisposes to colorectal cancer (CRC) with some specific features that distinguish it from sporadic CRC. Magnesium (Mg) homeostasis is severely compromised in IBD patients, which may affect both inflammation and tumor development. Efficient transcellular Mg transport in intestinal cells depends on the transient receptor potential melastatin (TRPM) channels type 6 and 7, but their expression has never been investigated in the context of IBD-related CRC. AIMS: We sought to study the expression pattern of TRPM6 and TRPM7 in CRC, and to compare IBD-related cases to sporadic cases. METHODS: TRPM6 and TRPM7 protein expression was evaluated by immunohistochemistry in surgical specimens from 16 IBD and 13 NON-IBD CRC patients. RESULTS: TRPM7 expression was higher in tumor tissue than in the adjacent non-neoplastic tissue in both IBD and NON-IBD patients. Overall, adenocarcinomas showed a higher TRPM7 expression than adenomas. TRPM7 expression also positively correlated with tumor grade. Conversely, TRPM6 expression was higher in tumor tissues in both IBD and NON-IBD CRC, but it did not correlate with tumor stage or grade. CONCLUSIONS: We report a possible participation of TRPM6 and 7 in both IBD-related and sporadic CRC and suggest that TRPM7 might serve as a marker of malignant transformation and lack of differentiation.


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Doenças Inflamatórias Intestinais/genética , Proteínas Serina-Treonina Quinases/genética , Canais de Cátion TRPM/genética , Adenocarcinoma/etiologia , Biomarcadores/sangue , Estudos de Casos e Controles , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/patologia , Feminino , Expressão Gênica , Humanos , Doenças Inflamatórias Intestinais/complicações , Magnésio/metabolismo , Masculino
19.
Nutr Cancer ; 61(1): 131-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19116883

RESUMO

Low Mg availability reversibly inhibited the growth of mammary epithelial HC11 cells by increasing the number of cells in the G0/G1 phase of the cell cycle. Because low Mg has been reported to promote oxidative reactions, we considered that low Mg-dependent growth arrest was mediated by oxidative stress. Surprisingly, both dichlorofluorescein-detectable reactive oxygen species and hydrogen peroxide-induced oxidative DNA damage were found to be lower in cells cultured in low Mg than in cells grown under control or high-Mg conditions. Gene expression profiling of low- and high-Mg cells showed the modulation of several genes, some regulating cell proliferation. In addition, low Mg cells also displayed overexpression of glutathione S-transferase (GST), leading to increased enzymatic activity. Of note, GST has been shown to modulate cell growth; therefore, we suggest that in low-Mg cells, GST upregulation might have a dual role in protecting against oxidative stress and in modulating cell proliferation.


Assuntos
Divisão Celular/efeitos dos fármacos , Células Epiteliais/citologia , Deficiência de Magnésio/patologia , Magnésio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Ensaio Cometa , Dano ao DNA , Relação Dose-Resposta a Droga , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Perfilação da Expressão Gênica , Glutationa Transferase/metabolismo , Humanos , Deficiência de Magnésio/fisiopatologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/fisiologia , Regulação para Cima
20.
Magnes Res ; 22(1): 5-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19441269

RESUMO

Over recent decades there have been several papers that documented hypomagnesaemia*, in cancer patients treated, with cisplatin, with combined chemotherapies and more recently, with cetuximab an antibody against the epidermal growth factor receptor. Recently, however, the clinical read-out of cetuximab-induced hypomagnesaemia has received different interpretations. Some reports called the readers' attention to the importance of magnesium supplementation in relieving patient's discomfort or preventing the most severe complications of hypomagnesaemia. Other reports claimed that magnesium deficiency could contribute to the oncologic efficacy of cetuximab. This latter interpretation implies that the decision on magnesium supplementation should consider the pros and cons of returning magnesium to normal levels. Given that decreased magnesium availability inhibits cell proliferation, hypomagnesaemia may slow down tumor growth. Unfortunately, one view does not fit all. We think it important to recapitulate our knowledge on the effects of magnesium on tumor growth, angiogenesis, invasion and metastatization with the aim of providing clinical oncologists with an overview of available data of the potential effects of hypomagnesaemia in tumor growth. Translating these results into clinical settings may help in designing suitable studies to better evaluate the consequences of hypomagnesaemia in cancer patients.


Assuntos
Antineoplásicos/efeitos adversos , Deficiência de Magnésio/induzido quimicamente , Magnésio/sangue , Neoplasias/sangue , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados , Cetuximab , Humanos , Deficiência de Magnésio/tratamento farmacológico , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA