Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Clin Nutr ESPEN ; 57: 501-509, 2023 10.
Article in English | MEDLINE | ID: mdl-37739699

ABSTRACT

BACKGROUND AND AIMS: The beneficial effect of low-glycemic index (GI) diet on gestational diabetes mellitus (GDM) has been suggested in many observational studies; however, results from intervention trials remain inconsistent. This study aims to estimate the effect of interventions with low-GI dietary advice on pregnant outcomes in women with elevated risk of GDM. DESIGN: PubMed, Web of Science, Embase, and Cochrane Library databases were searched for randomized clinical trials (RCTs) through March 2022. Studies reporting the effect of low-GI diet advice intervention on maternal and fetal outcomes in pregnant women with increased risks of GDM were included. Random or fixed effects model was used to calculate combined treatment effects. Publication bias was assessed via Begg's and Egger's tests and funnel plot inspection. RESULTS: Nine RCTs recruiting 3416 participants were included. Low-GI diet advice did not modulate the risk of GDM. Compared with control diets, low-GI diet advice significantly reduced gestational weight gain (GWG) (weighted mean differences, WMD = -0.93 kg, 95% CI: -1.31, -0.55; p < 0.001; n = 7) and the risk of premature birth (RR = 0.55, 95% CI: 0.35, 0.88; p = 0.012; n = 5). In subgroup analyses, the effect of low-GI diet interventions on premature birth was significant only in women with BMI higher than 30 kg/m2 (RR = 0.28, 95% CI: 0.10, 0.77, p = 0.014; n = 3); the significant effect on GWG was not altered by stratification of BMI and the type of GDM risk factors. No significant changes in other maternal and newborn outcomes were found. CONCLUSIONS: Low-GI diet advice interventions during pregnancy decreased GWG and the risk of premature birth in women with elevated GDM risk; however, the interventions did not significantly prevent GDM development in these women.


Subject(s)
Diabetes, Gestational , Premature Birth , Female , Humans , Infant, Newborn , Pregnancy , Combined Modality Therapy , Databases, Factual , Glycemic Index , Randomized Controlled Trials as Topic
2.
Adv Sci (Weinh) ; 10(13): e2205436, 2023 05.
Article in English | MEDLINE | ID: mdl-36840630

ABSTRACT

Ferroptosis has been linked to the pathogenesis of hepatic injury induced by ischemia/reperfusion (I/R). However, the mechanistic basis remains unclear. In this study, by using a mouse model of hepatic I/R injury, it is observed that glutathione (GSH) and cysteine depletion are associated with deficiency of the reducing power of nicotinamide adenine dinucleotide phosphate (NADPH). Genes involved in maintaining NADPH homeostasis are screened, and it is identified that I/R-induced hepatic ferroptosis is significantly associated with reduced expression and activity of NADP+ -dependent malic enzyme 1 (Me1). Mice with hepatocyte-specific Me1 gene deletion exhibit aggravated ferroptosis and liver injury under I/R treatment; while supplementation with L-malate, the substrate of ME1, restores NADPH and GSH levels and eventually inhibits I/R-induced hepatic ferroptosis and injury. A mechanistic study further reveals that downregulation of hepatic Me1 expression is largely mediated by the phosphatase and tensin homologue (PTEN)-dependent suppression of the mechanistic target of rapamycin/sterol regulatory element-binding protein 1 (mTOR/SREBP1) signaling pathway in hepatic I/R model. Finally, PTEN inhibitor, mTOR activator, or SREBP1 over-expression all increase hepatic NADPH, block ferroptosis, and protect liver against I/R injury. Taken together, the findings suggest that targeting ME1 may provide new therapeutic opportunities for I/R injury and other ferroptosis-related hepatic conditions.


Subject(s)
Liver , Reperfusion Injury , Humans , NADP/metabolism , Liver/metabolism , TOR Serine-Threonine Kinases/metabolism , Reperfusion Injury/pathology , Glutathione/metabolism
3.
Nat Rev Cardiol ; 20(1): 7-23, 2023 01.
Article in English | MEDLINE | ID: mdl-35788564

ABSTRACT

The maintenance of iron homeostasis is essential for proper cardiac function. A growing body of evidence suggests that iron imbalance is the common denominator in many subtypes of cardiovascular disease. In the past 10 years, ferroptosis, an iron-dependent form of regulated cell death, has become increasingly recognized as an important process that mediates the pathogenesis and progression of numerous cardiovascular diseases, including atherosclerosis, drug-induced heart failure, myocardial ischaemia-reperfusion injury, sepsis-induced cardiomyopathy, arrhythmia and diabetic cardiomyopathy. Therefore, a thorough understanding of the mechanisms involved in the regulation of iron metabolism and ferroptosis in cardiomyocytes might lead to improvements in disease management. In this Review, we summarize the relationship between the metabolic and molecular pathways of iron signalling and ferroptosis in the context of cardiovascular disease. We also discuss the potential targets of ferroptosis in the treatment of cardiovascular disease and describe the current limitations and future directions of these novel treatment targets.


Subject(s)
Cardiovascular Diseases , Ferroptosis , Humans , Iron/metabolism , Cardiovascular Diseases/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction
4.
J Mol Cell Cardiol ; 174: 15-24, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36375319

ABSTRACT

Iron is necessary for the life of practically all living things, yet it may also harm people toxically. Accordingly, humans and other mammals have evolved an effective and tightly regulatory system to maintain iron homeostasis in healthy tissues, including the heart. Iron deficiency is common in patients with heart failure, and is associated with worse prognosis in this population; while the prevalence of iron overload-related cardiovascular disorders is also increasing. Therefore, enhancing the therapy of patients with cardiovascular disorders requires a thorough understanding of iron homeostasis. Here, we give readers an overview of the fundamental mechanisms governing systemic iron homeostasis as well as the most recent knowledge about the intake, storage, use, and export of iron from the heart. Genetic mouse models used for investigation of iron metabolism in various in vivo scenarios are summarized and highlighted. We also go through different clinical conditions and therapeutic approaches that target cardiac iron dyshomeostasis. Finally, we conclude the review by outlining the present knowledge gaps and important open questions in this field in order to guide future research on cardiac iron metabolism.


Subject(s)
Heart Failure , Iron Deficiencies , Humans , Animals , Mice , Iron/metabolism , Heart , Heart Failure/metabolism , Homeostasis , Mammals/metabolism
5.
Alzheimers Res Ther ; 14(1): 184, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36514123

ABSTRACT

BACKGROUND: Growing evidence has showed an association between habitual glucosamine use and type 2 diabetes (T2D). However, the effect of habitual glucosamine use on risk of dementia remains poorly understood. Our study aimed to examine the association between glucosamine use and risk of dementia and further to identify the mediating role of T2D in the association. METHODS: A total of 495,942 participants from UK Biobank who completed a questionnaire on habitual glucosamine use were included at baseline (2006-2010) and then followed up for incidence of dementia until 2020. Cox proportional hazard regressions were performed to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for incident dementia. Markov multi-state models were used to explore the role of incidence of T2D during the follow-up in the association. RESULTS: Overall, 18.80% of the participants reported habitual use of glucosamine at baseline. A total of 6831 dementia events were recorded during a median follow-up of 11 years. In fully adjusted models, habitual glucosamine use was associated with a significantly lower risk of dementia (HR = 0.87, 95% CI: 0.82-0.93). Multi-state models showed that the association between glucosamine use and dementia was mediated by the incidence of T2D during the follow-up (HR of dementia without T2D: 0.92, 95% CI: 0.86-0.99; HR of post-T2D dementia: 0.79, 95% CI: 0.67-0.93). CONCLUSIONS: Our findings reveal that habitual use of glucosamine supplement is associated with a lower risk of dementia, which might be explained by incidence of T2D.


Subject(s)
Dementia , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/epidemiology , Glucosamine/therapeutic use , Risk Factors , Prospective Studies , Incidence , Dementia/epidemiology , Dementia/complications
6.
Front Nutr ; 9: 904957, 2022.
Article in English | MEDLINE | ID: mdl-35795586

ABSTRACT

Background: Epidemiological studies of citrus consumption in relation to melanoma risk have yielded conflicting results. This meta-analysis was performed to investigate the dose-response association between citrus consumption and risk of melanoma. Methods: Relevant prospective cohort studies were identified by searching PubMed, Embase, Scopus, and Web of Science databases up to February 28th, 2022. Results from individual studies were pooled using a random-effects model. Results: Five prospective studies, with 8,836 melanoma cases and 977,558 participants, were included in the meta-analysis. A significantly increased risk of melanoma was associated with the highest categories of either total citrus products (RR: 1.20; 95% CI: 1.01-1.42) or citrus fruit consumption (1.15; 1.04-1.28), but consumption of citrus juice was not associated with melanoma risk (1.08; 0.97-1.21). The dose-response analyses revealed that for per 1 serving/day increase in total citrus or citrus fruit consumption, the risk of melanoma increased by 9 and 12%, respectively. An inverted U-shaped curvilinear relationship, but not linear association, was observed between citrus juice consumption and melanoma risk. Conclusions: Citrus consumption was generally associated with a greater risk of malignant melanoma. Our findings may have important public health implications with respect to preventing melanoma.

7.
Food Funct ; 13(16): 8388-8398, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35856090

ABSTRACT

Background: The preventive effects of probiotic supplementation against gestational diabetes mellitus (GDM) in pregnant women remain unclear. The objective of this review was to investigate the effect of probiotic supplementation on the profiles of glucose metabolism in pregnant women without diabetes. The published literature was retrieved and screened from PubMed, Embase, Web of Science, CNKI (China National Knowledge Infrastructure), Wanfang, and Cochrane Center Register of Controlled Trails up to April 1st, 2021. Random controlled trials (RCTs) of probiotic supplementation on pregnant women without GDM were included. Results: 12 RCTs (2213 participants) were eligible for meta-analyses. Overall, probiotic supplementation significantly reduced GDM incidence (Risk Ratio (RR) = 0.62, 95% CI: 0.39-0.99), serum fasting blood glucose (FBG) (Mean Difference (MD) = -0.14 mmol L-1; 95% CI: -0.26 mmol L-1, -0.01 mmol L-1), insulin concentration (MD = -1.91 pmol L-1, 95% CI: -2.41 to -1.41), the homeostasis model assessment of insulin resistance (HOMA-IR) (MD = -0.32 mmol L-1; 95% CI: -0.42 mmol L-1, -0.22 mmol L-1), and Quantitative Insulin sensitivity Check Index (QUICKI) (MD = 0.02, 95% CI: 0.01,0.03) in pregnant women. Probiotic supplementation had no significant effects on the results of the oral glucose tolerance test (OGTT) (1 h OGTT, MD = -0.10, 95% CI: -0.30, 0.09; 2 h OGTT, MD = -0.06, 95% CI: -0.31, 0.20). Conclusion: This meta-analysis suggested that probiotic supplementation may lead to an improvement in glycemic control and reduction of GDM incidence in pregnant women.


Subject(s)
Diabetes, Gestational , Insulin Resistance , Probiotics , Blood Glucose/metabolism , Diabetes, Gestational/metabolism , Female , Humans , Pregnancy , Pregnant Women
8.
Front Nutr ; 9: 1084860, 2022.
Article in English | MEDLINE | ID: mdl-36590208

ABSTRACT

Background: Mental disorders account for an enormous global burden of disease, and has been associated with disturbed iron metabolism in observational studies. However, such associations are inconsistent and may be attributable to confounding from environmental factors. This study uses a two-sample Mendelian randomization (MR) analysis to investigate whether there is any causal effect of systemic iron status on risk of 24 specific mental disorders. Methods: Genetic variants with concordant relations to 4 biomarkers of iron status (serum iron, ferritin, transferrin saturation, and transferrin) were obtained from a genome-wide association study performed by the Genetics of Iron Status (GIS) consortium. Summary-level data for mental disorders were obtained from the UK Biobank. An inverse-variance weighted (IVW) approach was used for the main analysis, and the simple median, weighted median and MR-Egger methods were used in sensitivity analyses. Results: Genetically predicted serum iron, ferritin, and transferrin saturation were positively associated with depression and psychogenic disorder, and inversely associated with gender identity disorders. A higher transferrin, indicative of lower iron status, was also associated with increased risk of gender identity disorders and decreased risk of psychogenic disorder. Results were broadly consistent when using multiple sensitivity analyses to account for potential genetic pleiotropy. Conclusion: Our findings offer a novel insight into mental health, highlighting a detrimental effect of higher iron status on depression and psychogenic disorder as well as a potential protective role on risk of gender identity disorders. Further studies regarding the underlying mechanisms are warranted for updating preventative strategies.

9.
J Clin Endocrinol Metab ; 106(11): e4641-e4651, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34147035

ABSTRACT

CONTEXT: Iron overload is a known risk factor for type 2 diabetes (T2D); however, iron overload and iron deficiency have both been associated with metabolic disorders in observational studies. OBJECTIVE: Using mendelian randomization (MR), we assessed how genetically predicted systemic iron status affected T2D risk. METHODS: A 2-sample MR analysis was used to obtain a causal estimate. We selected genetic variants strongly associated (P < 5 × 10-8) with 4 biomarkers of systemic iron status from a study involving 48 972 individuals performed by the Genetics of Iron Status consortium and applied these biomarkers to the T2D case-control study (74 124 cases and 824 006 controls) performed by the Diabetes Genetics Replication and Meta-analysis consortium. The simple median, weighted median, MR-Egger, MR analysis using mixture-model, weighted allele scores, and MR based on a Bayesian model averaging approaches were used for the sensitivity analysis. RESULTS: Genetically instrumented serum iron (odds ratio [OR]: 1.07; 95% CI, 1.02-1.12), ferritin (OR: 1.19; 95% CI, 1.08-1.32), and transferrin saturation (OR: 1.06; 95% CI, 1.02-1.09) were positively associated with T2D. In contrast, genetically instrumented transferrin, a marker of reduced iron status, was inversely associated with T2D (OR: 0.91; 95% CI, 0.87-0.96). CONCLUSION: Genetic evidence supports a causal link between increased systemic iron status and increased T2D risk. Further studies involving various ethnic backgrounds based on individual-level data and studies regarding the underlying mechanism are warranted for reducing the risk of T2D.


Subject(s)
Biomarkers/metabolism , Diabetes Mellitus, Type 2/pathology , Genetic Predisposition to Disease , Iron Overload/complications , Iron/metabolism , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Case-Control Studies , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Female , Follow-Up Studies , Genome-Wide Association Study , Humans , Male , Prognosis , Risk Factors
10.
Int J Cancer ; 149(7): 1421-1425, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34004046

ABSTRACT

In observational studies of children and adolescents, higher body weight has been associated with distinct disease outcomes, including cancer, in adulthood. Therefore, we performed a two-sample Mendelian randomization (MR) study to evaluate the causal effect of childhood obesity on long-term cancer risk. Single-nucleotide polymorphisms associated with higher childhood body mass index (BMI) from large-scale genome-wide association studies were used as genetic instruments. Summary-level data for 24 site-specific cancers were obtained from UK Biobank. We found that a 1-SD increase in childhood BMI (kg/m2 ) was significantly associated with a 60% increase in risk of pancreatic cancer (odds ratio [OR]: 1.60; 95% confidence interval [CI]: 1.12-2.28; P < 0.01) and a 47% increase in risk of esophageal cancer (OR: 1.47; 95% CI: 1.09-1.97; P < 0.01) in adults. In contrast, there was an inverse association of genetic predisposition to childhood obesity with throat (OR: 0.46; 95% CI: 0.27-0.79; P < 0.01) and breast cancer (OR: 0.77; 95% CI: 0.64-0.94; P < 0.01) in adult life. For the other 20 cancers studied, no statistically significant association was observed. Our MR analyses found causal effects of childhood obesity on several cancers. Maintaining a healthy weight should be emphasized during childhood and adolescence to prevent cancer risk later in life.


Subject(s)
Body Mass Index , Causality , Genetic Predisposition to Disease , Mendelian Randomization Analysis , Neoplasms/epidemiology , Pediatric Obesity/physiopathology , Adolescent , Child , Genome-Wide Association Study , Humans , Neoplasms/pathology , Prognosis , Risk Factors , United Kingdom/epidemiology
11.
J Pineal Res ; 70(2): e12704, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33206394

ABSTRACT

Accumulating evidence demonstrates that ferroptosis may be important in the pathophysiological process of traumatic brain injury (TBI). As a major hormone of the pineal gland, melatonin exerts many beneficial effects on TBI, but there is no information regarding the effects of melatonin on ferroptosis after TBI. As expected, TBI resulted in the time-course changes of ferroptosis-related molecules expression and iron accumulation in the ipsilateral cortex. Importantly, we found that treating with melatonin potently rescued TBI induced the changes mentioned above and improved functional deficits versus vehicle. Similar results were obtained with a ferroptosis inhibitor, liproxstatin-1. Moreover, the protective effect of melatonin is likely dependent on melatonin receptor 1B (MT2). Although ferritin plays a vital role in iron metabolism by storing excess cellular iron, its precise function in the brain, and whether it involves melatonin's neuroprotection remain unexplored. Considering ferritin H (Fth) is expressed predominantly in the neurons and global loss of Fth in mice induces early embryonic lethality, we then generated neuron-specific Fth conditional knockout (Fth-KO) mice, which are viable and fertile but have altered iron metabolism. In addition, Fth-KO mice were more susceptible to ferroptosis after TBI, and the neuroprotection by melatonin was largely abolished in Fth-KO mice. In vitro siFth experiments further confirmed the results mentioned above. Taken together, these data indicate that melatonin produces cerebroprotection, at least partly by inhibiting neuronal Fth-mediated ferroptosis following TBI, supporting the notion that melatonin is an excellent ferroptosis inhibitor and its anti-ferroptosis provides a potential therapeutic target for treating TBI.


Subject(s)
Apoferritins/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Melatonin/therapeutic use , Animals , Apoferritins/genetics , Blotting, Western , Ferroptosis/drug effects , Immunohistochemistry , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction
12.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 49(1): 58-70, 2020 05 25.
Article in Chinese | MEDLINE | ID: mdl-32621410

ABSTRACT

Iron homeostasis plays an important role for the maintenance of human health. It is known that iron metabolism is tightly regulated by several key genes, including divalent metal transport-1(DMT1), transferrin receptor 1(TFR1), transferrin receptor 2(TFR2), ferroportin(FPN), hepcidin(HAMP), hemojuvelin(HJV) and Ferritin H. Recently, it is reported that DNA methylation, histone acetylation, and microRNA (miRNA) epigenetically regulated iron homeostasis. Among these epigenetic regulators, DNA hypermethylation of the promoter region of FPN, TFR2, HAMP, HJV and bone morphogenetic protein 6 (BMP6) genes result in inhibitory effect on the expression of these iron-related gene. In addition, histone deacetylase (HADC) suppresses HAMP gene expression. On the contrary, HADC inhibitor upregulates HAMP gene expression. Additional reports showed that miRNA can also modulate iron absorption, transport, storage and utilization via downregulation of DMT1, FPN, TFR1, TFR2, Ferritin H and other genes. It is noteworthy that some key epigenetic regulatory enzymes, such as DNA demethylase TET2 and histone lysine demethylase JmjC KDMs, require iron for the enzymatic activities. In this review, we summarize the recent progress of DNA methylation, histone acetylation and miRNA in regulating iron metabolism and also discuss the future research directions.


Subject(s)
Epigenesis, Genetic , Homeostasis , Iron , Gene Expression Regulation/genetics , Humans , Iron/metabolism , Receptors, Transferrin
13.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 49(1): 44-57, 2020 05 25.
Article in Chinese | MEDLINE | ID: mdl-32621416

ABSTRACT

Recently, ferroptosis, an iron-dependent novel type of cell death, has been characterized as an excessive accumulation of lipid peroxides and reactive oxygen species. Emerging studies demonstrate that ferroptosis not only plays an important role in the pathogenesis and progression of chronic diseases, but also functions differently in the different disease context. Notably, it is shown that activation of ferroptosis could potently inhibit tumor growth and increase sensitivity to chemotherapy and immunotherapy in various cancer settings. As a result, the development of more efficacious ferroptosis agonists remains the mainstay of ferroptosis-targeting strategy for cancer therapeutics. By contrast, in non-cancerous chronic diseases, including cardiovascular & cerebrovascular diseases and neurodegenerative diseases, ferroptosis functions as a risk factor to promote these diseases progression through triggering or accelerating tissue injury. As a matter of fact, blocking ferroptosis has been demonstrated to effectively prevent ischemia-reperfusion heart disease in preclinical animal models. Therefore, it is a promising field to develope potent ferroptosis inhibitors for preventing and treating cardiovascular & cerebrovascular diseases and neurodegenerative diseases. In this article, we summarize the most recent progress on ferroptosis in chronic diseases, and draw attention to the possible clinical impact of this recently emerged ferroptosis modalities.


Subject(s)
Chronic Disease , Ferroptosis , Iron , Animals , Ferroptosis/physiology , Iron/metabolism , Reactive Oxygen Species
14.
Adv Nutr ; 11(6): 1510-1528, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32503038

ABSTRACT

Observational studies regarding the putative associations between dietary intake of homocysteine metabolism-related B-vitamins (vitamin B-6, folate, and vitamin B-12) and stroke risk have yielded inconsistent results. Thus, we conducted a systematic meta-analysis of prospective studies in order to examine the relation between the dietary (from diet and supplements) intake of these B-vitamins and the risk of stroke. PubMed and Web of Science were searched for relevant articles published through to 25 February, 2020, and RR of stroke in relation to dietary intake of vitamin B-6, folate, and vitamin B-12 were pooled using a random-effects model. Eleven publications of 12 prospective studies comprising 389,938 participants and 10,749 cases were included in the final analysis. We found that dietary intake of vitamin B-6 and folate were associated with a reduced risk of stroke, and this inverse association remained significant in studies with >10 y of follow-up periods and among participants without a pre-existing stroke event. A dose-response analysis revealed a linear inverse association between folate and vitamin B-6 intake and the risk of stroke, with a pooled RR of 0.94 (95% CI: 0.90-0.98) and 0.94 (95% CI: 0.89-0.99) for each 100 µg/d increment in folate intake and 0.5 mg/d increment in vitamin B-6 intake, respectively. In contrast, we found no significant association between dietary vitamin B-12 intake and the risk of stroke, with an RR of 1.01 (95% CI: 0.97-1.06) per 3 µg/d increase. In conclusion, our findings suggest that increased intake of vitamin B-6 and folate is associated with a reduced risk of stroke, supporting the notion that increasing habitual folate and vitamin B-6 intake may provide a small but beneficial effect with respect to stroke.


Subject(s)
Stroke , Adult , Aged , China , Eating , Female , Folic Acid , Follow-Up Studies , Homocysteine , Humans , Male , Middle Aged , Nutrition Surveys , Prospective Studies , Risk Factors , Stroke/epidemiology , Vitamins
15.
Research (Wash D C) ; 2020: 2402961, 2020.
Article in English | MEDLINE | ID: mdl-32377638

ABSTRACT

The recent outbreak of COVID-19 has been rapidly spreading on a global scale. To date, there is no specific vaccine against the causative virus, SARS-CoV-2, nor is there an effective medicine for treating COVID-19, thus raising concerns with respect to the effect of risk factors such as clinical course and pathophysiological parameters on disease severity and outcome in patients with COVID-19. By extracting and analyzing all available published clinical data, we identified several major clinical characteristics associated with increased disease severity and mortality among patients with COVID-19. Specifically, preexisting chronic conditions such as hypertension, cardiovascular disease, chronic kidney disease, and diabetes are strongly associated with an increased risk of developing severe COVID-19; surprisingly, however, we found no correlation between chronic liver disease and increased disease severity. In addition, we found that both acute cardiac injury and acute kidney injury are highly correlated with an increased risk of COVID-19-related mortality. Given the high risk of comorbidity and the high mortality rate associated with tissue damage, organ function should be monitored closely in patients diagnosed with COVID-19, and this approach should be included when establishing new guidelines for managing these high-risk patients. Moreover, additional clinical data are needed in order to determine whether a supportive therapy can help mitigate the development of severe, potentially fatal complications, and further studies are needed to identify the pathophysiology and the mechanism underlying this novel coronavirus-associated infectious disease. Taken together, these findings provide new insights regarding clinical strategies for improving the management and outcome of patients with COVID-19.

16.
Blood ; 136(6): 726-739, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32374849

ABSTRACT

Although the serum-abundant metal-binding protein transferrin (encoded by the Trf gene) is synthesized primarily in the liver, its function in the liver is largely unknown. Here, we generated hepatocyte-specific Trf knockout mice (Trf-LKO), which are viable and fertile but have impaired erythropoiesis and altered iron metabolism. Moreover, feeding Trf-LKO mice a high-iron diet increased their susceptibility to developing ferroptosis-induced liver fibrosis. Importantly, we found that treating Trf-LKO mice with the ferroptosis inhibitor ferrostatin-1 potently rescued liver fibrosis induced by either high dietary iron or carbon tetrachloride (CCl4) injections. In addition, deleting hepatic Slc39a14 expression in Trf-LKO mice significantly reduced hepatic iron accumulation, thereby reducing ferroptosis-mediated liver fibrosis induced by either a high-iron diet or CCl4 injections. Finally, we found that patients with liver cirrhosis have significantly lower levels of serum transferrin and hepatic transferrin, as well as higher levels of hepatic iron and lipid peroxidation, compared with healthy control subjects. Taken together, these data indicate that hepatic transferrin plays a protective role in maintaining liver function, providing a possible therapeutic target for preventing ferroptosis-induced liver fibrosis.


Subject(s)
Ferroptosis/physiology , Iron/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Transferrin/physiology , Animals , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cyclohexylamines/pharmacology , Cytokines/analysis , Erythropoiesis/physiology , Erythropoietin/analysis , Female , Ferroptosis/drug effects , Hepatocytes/metabolism , Homeostasis , Iron Overload/complications , Iron, Dietary/toxicity , Lipid Peroxidation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/analysis , Phenylenediamines/pharmacology , Transferrin/analysis
17.
Circ Res ; 127(4): 486-501, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32349646

ABSTRACT

RATIONALE: Maintaining iron homeostasis is essential for proper cardiac function. Both iron deficiency and iron overload are associated with cardiomyopathy and heart failure via complex mechanisms. Although ferritin plays a central role in iron metabolism by storing excess cellular iron, the molecular function of ferritin in cardiomyocytes remains unknown. OBJECTIVE: To characterize the functional role of Fth (ferritin H) in mediating cardiac iron homeostasis and heart disease. METHODS AND RESULTS: Mice expressing a conditional Fth knockout allele were crossed with 2 distinct Cre recombinase-expressing mouse lines, resulting in offspring that lack Fth expression specifically in myocytes (MCK-Cre) or cardiomyocytes (Myh6-Cre). Mice lacking Fth in cardiomyocytes had decreased cardiac iron levels and increased oxidative stress, resulting in mild cardiac injury upon aging. However, feeding these mice a high-iron diet caused severe cardiac injury and hypertrophic cardiomyopathy, with molecular features typical of ferroptosis, including reduced glutathione (GSH) levels and increased lipid peroxidation. Ferrostatin-1, a specific inhibitor of ferroptosis, rescued this phenotype, supporting the notion that ferroptosis plays a pathophysiological role in the heart. Finally, we found that Fth-deficient cardiomyocytes have reduced expression of the ferroptosis regulator Slc7a11, and overexpressing Slc7a11 selectively in cardiomyocytes increased GSH levels and prevented cardiac ferroptosis. CONCLUSIONS: Our findings provide compelling evidence that ferritin plays a major role in protecting against cardiac ferroptosis and subsequent heart failure, thereby providing a possible new therapeutic target for patients at risk of developing cardiomyopathy.


Subject(s)
Amino Acid Transport System y+/metabolism , Apoferritins/deficiency , Cardiomyopathies/etiology , Ferroptosis/physiology , Iron/metabolism , Myocardium/metabolism , Aging , Alleles , Animals , Apoferritins/adverse effects , Apoferritins/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/prevention & control , Crosses, Genetic , Cyclohexylamines/administration & dosage , Glutathione/metabolism , Heart Failure/etiology , Homeostasis , Hypertrophy, Left Ventricular/etiology , Iron Deficiencies , Iron Overload , Iron, Dietary/adverse effects , Lipid Peroxidation , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Oxidative Stress , Phenylenediamines/administration & dosage , Reactive Oxygen Species/metabolism
19.
Sci Rep ; 9(1): 19046, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836761

ABSTRACT

Growing evidence has suggested a possible relationship between dietary calcium intake and metabolic syndrome (MetS) risk. However, the findings of these observational studies are inconclusive, and the dose-response association between calcium intake and risk of MetS remains to be determined. Here, we identified relevant studies by searching PubMed and Web of Science databases up to December 2018, and selected observational studies reporting relative risk (RR) with 95% confidence interval (CI) for MetS based on calcium intake and estimated the summary RRs using random-effects models. Eight cross-sectional and two prospective cohort studies totaling 63,017 participants with 14,906 MetS cases were identified. A significantly reduced risk of MetS was associated with the highest levels of dietary calcium intake (RR: 0.89; 95% CI: 0.80-0.99; I2 = 75.3%), with stronger association and less heterogeneity among women (RR: 0.74, 95% CI: 0.66-0.83; I2 = 0.0%) than among men (RR: 1.06, 95% CI: 0.82-1.37; I2 = 72.6%). Our dose-response analysis revealed that for each 300 mg/day increase in calcium intake, the risk of MetS decreased by 7% (RR: 0.93; 95% CI: 0.87-0.99; I2 = 77.7%). In conclusion, our findings suggest that dietary calcium intake may be inversely associated with the risk of MetS. These findings may have important public health implications with respect to preventing the disease. Further studies, in particular longitudinal cohort studies and randomized clinical trials, will be necessary to determine whether calcium supplementation is effective to prevent MetS.


Subject(s)
Calcium, Dietary/pharmacology , Feeding Behavior , Metabolic Syndrome/prevention & control , Adult , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Risk Factors , Young Adult
20.
Am J Clin Nutr ; 110(1): 76-90, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31161192

ABSTRACT

BACKGROUND: Although many studies have shown that low zinc status is associated with diabetes, the putative effects of zinc supplementation on glycemic control are inconclusive. OBJECTIVES: The aim of this meta-analysis of randomized controlled trials was to assess the effects of zinc supplementation in preventing and managing diabetes. METHODS: PubMed, Embase, and the Cochrane Library were searched for articles that were published through February 10, 2019 and contained estimates for the outcomes of interest. The pooled results were then analyzed with the use of a random-effects model. RESULTS: Thirty-two placebo-controlled interventions were extracted from 36 publications, involving a total of 1700 participants in 14 countries. Overall, compared with their respective control groups, the subjects in the zinc-supplementation group had a statistically significant reduction in fasting glucose [FG, weighted mean difference (WMD): -14.15 mg/dL; 95% CI: -17.36, -10.93 mg/dL], 2-h postprandial glucose (WMD: -36.85 mg/dL; 95% CI: -62.05, -11.65 mg/dL), fasting insulin (WMD: -1.82 mU/L; 95% CI: -3.10, -0.54 mU/L), homeostasis model assessment for insulin resistance (WMD: -0.73; 95% CI: -1.22, -0.24), glycated hemoglobin (WMD: -0.55%; 95% CI: -0.84, -0.27%), and high-sensitivity C-reactive protein (WMD: -1.31 mg/L; 95% CI: -2.05, -0.56 mg/L) concentrations. Moreover, subgroup analyses revealed that the effects of zinc supplementation on FG are significantly influenced by diabetic status and the formulation of the zinc supplement. CONCLUSIONS: Our analysis revealed that several key glycemic indicators are significantly reduced by zinc supplementation, particularly the FG in subjects with diabetes and in subjects who received an inorganic zinc supplement. Together, these findings support the notion that zinc supplementation may have clinical potential as an adjunct therapy for preventing or managing diabetes. This trial was registered at PROSPERO as CRD42018111838.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 2/prevention & control , Randomized Controlled Trials as Topic , Zinc/administration & dosage , C-Reactive Protein/analysis , Dietary Supplements , Fasting , Glycated Hemoglobin/analysis , Humans , Insulin/blood , Insulin Resistance , Placebos
SELECTION OF CITATIONS
SEARCH DETAIL
...