Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Magnes Res ; 0(0)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652733

ABSTRACT

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.

2.
Eur J Nutr ; 61(2): 625-636, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34687321

ABSTRACT

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.


Subject(s)
COVID-19 , Homeostasis , Humans , Magnesium , Pandemics , SARS-CoV-2
3.
Nutrients ; 13(12)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34959740

ABSTRACT

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.


Subject(s)
Colitis/microbiology , Colitis/therapy , Gastrointestinal Microbiome/drug effects , Magnesium/pharmacology , Animals , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Dysbiosis/microbiology , Dysbiosis/therapy , Feces/microbiology , Female , Magnesium Deficiency/microbiology , Magnesium Deficiency/therapy , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S
5.
Nutrients ; 12(11)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114586

ABSTRACT

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.


Subject(s)
Cetuximab/adverse effects , Epidermal Growth Factor/metabolism , Intestinal Absorption/drug effects , Magnesium/metabolism , TRPM Cation Channels/metabolism , Caco-2 Cells , Colon/metabolism , Humans , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/metabolism
6.
Cell Calcium ; 83: 102078, 2019 11.
Article in English | MEDLINE | ID: mdl-31493712

ABSTRACT

Derangement of magnesium homeostasis underlies the pathophysiology of many diseases, including cancer. Recent advances support the view that aberrant expression of Mg2+ channels and other Mg2+ homeostatic factors may affect many hallmarks of cancer. The seminal idea of magnesium as a key regulator of cell proliferation has been enriched by novel intriguing findings that link magnesium and Mg2+ transporters to distinctive and complementary capabilities that enable tumour growth and metastatic dissemination. In this review, we examine the evidence on the involvement of members from the TRPM, CNNM and SCL41 protein families in cancer progression, and discuss their potential as therapeutic targets.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Membrane/metabolism , Neoplasms/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Cell Proliferation , Homeostasis , Humans , Protein Conformation , Protein Domains/genetics , Structure-Activity Relationship
7.
Nutrients ; 10(6)2018 Jun 18.
Article in English | MEDLINE | ID: mdl-29912157

ABSTRACT

Intestinal magnesium (Mg) uptake is essential for systemic Mg homeostasis. Colon cells express the two highly homologous transient receptor potential melastatin type (TRPM) 6 and 7 Mg2+ channels, but their precise function and the consequences of their mutual interaction are not clear. To explore the functional role of TRPM6 and TRPM7 in the colon, we used human colon cell lines that innately express both channels and analyzed the functional consequences of genetic knocking-down, by RNA interference, or pharmacological inhibition, by NS8593, of either channel. TRPM7 silencing caused an increase in Mg2+ influx, and correspondingly enhanced cell proliferation and migration, while downregulation of TRPM6 did not affect significantly either Mg2+ influx or cell proliferation. Exposure to the specific TRPM6/7 inhibitor NS8593 reduced Mg2+ influx, and consequently cell proliferation and migration, but Mg supplementation rescued the inhibition. We propose a model whereby in colon cells the functional Mg2+ channel at the plasma membrane may consist of both TRPM7 homomers and TRPM6/7 heteromers. A different expression ratio between the two proteins may result in different functional properties. Altogether, our findings confirm that TRPM6 cannot be replaced by TRPM7, and that TRPM6/7 complexes and TRPM6/7-mediated Mg2+ influx are indispensable in human epithelial colon cells.


Subject(s)
Colon/cytology , Colon/metabolism , Epithelial Cells/physiology , Magnesium/metabolism , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Cell Line, Tumor , Humans , Intestinal Mucosa/cytology , Protein Serine-Threonine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , TRPM Cation Channels/genetics
8.
Inflamm Bowel Dis ; 24(10): 2198-2210, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29788266

ABSTRACT

Background: Magnesium (Mg) is essential for human health and is absorbed mainly in the intestine. In view of the likely occurrence of an Mg deficit in inflammatory bowel disease (IBD) and the documented role of Mg in modulating inflammation, the present study addresses whether Mg availability can affect the onset and progression of intestinal inflammation. Methods: To study the correlation between Mg status and disease activity, we measured magnesemia by atomic absorption spectroscopy in a cohort of IBD patients. The effects of dietary Mg modulation were assessed in a murine model of dextran sodium sulfate (DSS)-induced colitis by monitoring magnesemia, weight, fecal occult blood, diarrhea, colon length, and histology. Expression of the transient receptor potential melastatin (TRPM) 6 channel was assessed by real-time reverse transcription polymerase chain reaction and immunohistochemistry in murine colon tissues. The effect of Mg on epithelial barrier formation/repair was evaluated in human colon cell lines. Results: Inflammatory bowel disease patients presented with a substantial Mg deficit, and serum Mg levels were inversely correlated with disease activity. In mice, an Mg-deficient diet caused hypomagnesemia and aggravated DSS-induced colitis. Colitis severely compromised intestinal Mg2+ absorption due to mucosal damage and reduction in TRPM6 expression, but Mg supplementation resulted in better restoration of mucosal integrity and channel expression. Conclusions: Our results highlight the importance of evaluating and correcting magnesemia in IBD patients. The murine model suggests that Mg supplementation may represent a safe and cost-effective strategy to reduce inflammation and restore normal mucosal function.


Subject(s)
Colitis, Ulcerative/complications , Colitis/prevention & control , Crohn Disease/complications , Diet , Hypocalcemia/metabolism , Magnesium Deficiency/congenital , Magnesium/administration & dosage , TRPM Cation Channels/metabolism , Adult , Aged , Aged, 80 and over , Animals , Case-Control Studies , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/physiopathology , Crohn Disease/metabolism , Crohn Disease/physiopathology , Dextran Sulfate/toxicity , Female , Follow-Up Studies , Humans , Hypocalcemia/etiology , Hypocalcemia/pathology , Magnesium/metabolism , Magnesium Deficiency/etiology , Magnesium Deficiency/metabolism , Magnesium Deficiency/pathology , Male , Mice, Inbred C57BL , Middle Aged , Prognosis , TRPM Cation Channels/genetics , Young Adult
9.
Chem Res Toxicol ; 29(3): 317-22, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26918850

ABSTRACT

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.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/metabolism , Doxorubicin/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Magnesium/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship , TRPM Cation Channels/metabolism
10.
Magnes Res ; 28(2): 46-55, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26422833

ABSTRACT

Disturbances in magnesium homeostasis, often linked to altered expression and/or function of magnesium channels, have been implicated in a plethora of diseases. This review focuses on magnesium transporter 1 (MAGT1), as recently described changes in this gene have further extended our understanding of the role of magnesium in human health and disease. The identification of genetic changes and their functional consequences in patients with immunodeficiency revealed that magnesium and MAGT1 are key molecular players for T cell-mediated immune responses. This led to the description of XMEN (X-linked immunodeficiency with magnesium defect, Epstein Barr Virus infection, and neoplasia) syndrome, for which Mg2+ supplementation has been shown to be beneficial. Similarly, the identification of a copy-number variation (CNV) leading to dysfunctional MAGT1 in a family with atypical ATRX syndrome and skin abnormalities, suggested that the MAGT1 defect could be responsible for the cutaneous problems. On the other hand, recent genetic investigations question the previously proposed role for MAGT1 in intellectual disability. Understanding the molecular basis of the involvement of magnesium and its channels in human pathogenesis will improve opportunities for Mg2+ therapies in the clinic.


Subject(s)
Cation Transport Proteins/physiology , Immunologic Deficiency Syndromes/genetics , Intellectual Disability/genetics , Animals , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/metabolism , Intellectual Disability/diagnosis , Intellectual Disability/metabolism , Magnesium/metabolism , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/metabolism
11.
Magnes Res ; 24(3): S122-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21951649

ABSTRACT

The epithelial Mg(2+) channel TRPM6 is considered a pivotal component in active Mg(2+)absorption and re-absorption in the intestine and kidney, but its expression and function in other tissues are largely unknown. We have previously demonstrated that extracellular Mg(2+) availability modulates TRPM6, but not the ubiquitous TRPM7, in cultured mammary epithelial cells; in addition, TRPM6 protein expression correlated to Mg(2+) influx capacities. Our results closely remind the modulation of TRPM6 described by others in murine kidney and colon following Mg(2+) dietary restriction. We sought to validate our observations by investigating whether TRPM6 modulation by extracellular Mg(2+)also occurs in vivo. To this aim, we exploited a model consisting of rats fed either with a Mg(2+)-deficient or a Mg(2+)-enriched diet, and studied TRPM6 expression in breast and kidney tissues. Immunohistochemical and western blot analyses confirmed that rat mammary tissues express TRPM6 protein levels similar to those found in the kidney, and that protein expression is modulated by dietary Mg(2+). In particular, Mg(2+) restriction upregulated TRPM6 expression, while Mg(2+) supplementation resulted in a significant decrease in protein levels. This work confirms and extends our previous results on TRPM6 modulation by Mg(2+) availability in mammary tissues. Further studies are required to clarify the functional significance of these findings, and the role of TRPM6 in tissue-specific magnesium homeostasis.


Subject(s)
Dietary Supplements , Epithelial Cells/metabolism , Magnesium/pharmacology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , TRPM Cation Channels/metabolism , Animals , Blotting, Western , Body Weight/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Magnesium/blood , Mammary Glands, Animal/drug effects , Organ Size/drug effects , Rats , Rats, Sprague-Dawley
12.
Magnes Res ; 22(1): 5-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19441269

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/adverse effects , Magnesium Deficiency/chemically induced , Magnesium/blood , Neoplasms/blood , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Cetuximab , Humans , Magnesium Deficiency/drug therapy , Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL