Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
BMC Vet Res ; 20(1): 210, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762716

ABSTRACT

BACKGROUND: Myxomatous mitral valve disease (MMVD) is the most common acquired cardiovascular disease in small breed dogs. In contrast to human patients with heart failure (HF), iron deficiency (ID) prevalence in dogs with MMVD is weakly known. The study aimed to assess the usability of ID markers in serum and reticulocyte parameters from whole blood of dogs with MMVD to evaluate early ID symptoms. RESULTS: Sixty-eight dogs (43 male and 25 female) were included in the study. MMVD dogs were assigned according to the 2019 ACVIM guidelines for groups B1 (n = 9), B2 (n = 10), C (n = 27) and D (n = 10). Groups were also combined into B1 and B2 as non-symptomatic HF and C with D as symptomatic HF. Healthy controls were 12 dogs. Serum iron concentration below the reference range in dogs with MMVD was 12.5%. Other ID indices, such as %SAT, UIBC, and TIBC were similar in the MMVD groups and healthy controls (p > 0.05 for all parameters). Statistical comparison between control group and 4 groups of different stages of MMVD showed that significant differences occur only in serum transferrin. The assessment of ferritin and soluble transferrin receptors using Western Blotting did not show differences between control (n = 7) and MMVD (n = 33) dogs. Study has shown positive correlation between ID parameters and echocardiographic indices such as LA/Ao and LVIDdN, and some biochemical parameters. A significant increase in reticulocytes percentage, assessed manually, was observed in the HF group of animals (p = 0.027) compared to the control group. CONCLUSIONS: Studies have shown that ID parameters in serum are not significantly different in dogs with MMVD compared to healthy dogs. However, there is a clear correlation between atrial size and normalised left ventricular size to body size and some biochemical parameters, including ID parameters and therefore the severity of MMVD.


Subject(s)
Dog Diseases , Iron , Dogs , Animals , Dog Diseases/blood , Female , Male , Iron/blood , Biomarkers/blood , Ferritins/blood , Mitral Valve Insufficiency/veterinary , Mitral Valve Insufficiency/blood , Iron Deficiencies/blood , Heart Valve Diseases/veterinary , Heart Valve Diseases/blood , Mitral Valve , Anemia, Iron-Deficiency/veterinary , Anemia, Iron-Deficiency/blood , Transferrin/analysis , Transferrin/metabolism , Reticulocytes
2.
Cardiol J ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37853824

ABSTRACT

BACKGROUND: Skeletal muscle dysfunction is a feature of heart failure (HF). Iron deficiency (ID) is prevalent in patients with HF associated with exercise intolerance and poor quality of life. Intravenous iron in iron deficient patients with HF has attenuated HF symptoms, however the pathomechanisms remain unclear. The aim of study was to assess whether intravenous iron supplementation as compared to placebo improves energy metabolism of skeletal muscles in patients with HF. METHODS: Men with heart failure with reduced ejection fraction (HFrEF) and ID were randomised in 1:1 ratio to either intravenous ferric carboxymaltose (IV FCM) or placebo. In vivo reduction of lactates by exercising skeletal muscles of forearm was analyzed. A change in lactate production between week 0 and 24 was considered as a primary endpoint of the study. RESULTS: There were two study arms: the placebo and the IV FCM (12 and 11 male patients with HFrEF). At baseline, there were no differences between these two study arms. IV FCM therapy as compared to placebo reduced the exertional production of lactates in exercising skeletal muscles. These effects were accompanied by a significant increase in both serum ferritin and transferrin saturation in the IV FCM arm which was not demonstrated in the placebo arm. CONCLUSIONS: Intravenous iron supplementation in iron deficient men with HFrEF improves the functioning of skeletal muscles via an improvement in energy metabolism in exercising skeletal muscles, limiting the contribution of anaerobic reactions generating ATP as reflected by a lower in vivo lactate production in exercising muscles in patients with repleted iron stores.

3.
Biomedicines ; 11(8)2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37626633

ABSTRACT

Pathomechanisms responsible for recovery from acute myocarditis (MCD) or progression to non-ischemic cardiomyopathy have not been comprehensively investigated. Iron, positioned at the crossroads of inflammation and the energy metabolism of cardiomyocytes, may contribute to the pathophysiology of inflammatory myocardial disease. The aim of this study was to evaluate whether systemic iron parameters are related to myocardial dysfunction in MCD patients. We prospectively enrolled 42 consecutive patients hospitalized for MCD. Their iron status and their clinical, laboratory, and echocardiographic indices were assessed during hospitalization and during ambulatory visits six weeks after discharge. A control group comprising healthy volunteers was recruited. The MCD patients had higher serum ferritin and hepcidin and lower serum iron concentration and transferrin saturation (TSAT) than the healthy controls (all p < 0.01). Six weeks after discharge, the iron status of the MCD patients was already comparable to that of the control group. During hospitalization, lower serum iron and TSAT correlated with higher NT-proBNP (both p < 0.05). In-hospital lower serum iron and TSAT correlated with both a lower left ventricular ejection fraction (LVEF) and worse left ventricular global longitudinal strain at follow-up visits (all p < 0.05). In conclusion, in patients with acute MCD, iron status is altered and normalizes within six weeks. Low serum iron and TSAT are related to greater in-hospital neurohormonal activation and subtle persistent left ventricular dysfunction.

4.
Antioxidants (Basel) ; 12(6)2023 May 25.
Article in English | MEDLINE | ID: mdl-37371882

ABSTRACT

Metabolic syndrome (MetS) significantly increases the risk of cardiovascular diseases (CVD), a leading cause of death globally. The presented study investigated the cardioprotective role of dietary polyphenols found in pomegranate peels in an animal model of metabolic syndrome. Zucker diabetic fatty rats (ZDF, MetS rats, fa/fa) were supplemented with polyphenol-rich pomegranate peel extract (EPP) at two dosages: 100 mg/kg BW and 200 mg/kg BW. The extract was administered for 8 weeks. The effect of ethanolic peel extract on the concentration of oxidative stress markers (CAT, SOD, MnSOD, GR, GST, GPx, TOS, SH, and MDA), biomarkers of heart failure (cTnI, GAL-3), and alternations in tissue architecture was assessed. The results showed a significant increase in SH concentration mediated via EPP supplementation (p < 0.001). Treatment with a 100 mg/kg BW dosage reduced the TOS level more efficiently than the higher dose. Interestingly, the CAT and GST activities were relevantly higher in the MetS 100 group (p < 0.001) compared to the MetS control. The rats administered EPP at a dose of 200 mg/kg BW did not follow a similar trend. No differences in the GR (p = 0.063), SOD (p = 0.455), MnSOD (p = 0.155), and MDA (p = 0.790) concentration were observed after exposure to the pomegranate peel extract. The administration of EPP did not influence the cTnI and GAL-3 levels. Histology analysis of the heart and aorta sections revealed no toxic changes in phenolic-treated rats. The findings of this study prove that the extract from pomegranate peels possesses free radical scavenging properties in the myocardium. The effect on alleviating ventricular remodeling and cardiomyocyte necrosis was not confirmed and requires further investigation.

5.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162949

ABSTRACT

Despite advances in the management of iron deficiency in heart failure (HF), the mechanisms underlying the effects of treatment remain to be established. Iron distribution and metabolism in HF pathogenesis need to be clarified. We used a porcine tachycardia-induced cardiomyopathy model to find out how HF development influences hepatic and myocardial iron storing, focusing on ferritin, the main iron storage protein. We found that cumulative liver congestion (due to the decrease of heart function) overwhelms its capacity to recycle iron from erythrocytes. As a consequence, iron is trapped in the liver as poorly mobilized hemosiderin. What is more, the ferritin-bound Fe3+ (reflecting bioavailable iron stores), and assembled ferritin (reflecting ability to store iron) are decreased in HF progression in the liver. We demonstrate that while HF pigs show iron deficiency indices, erythropoiesis is enhanced. Renin-angiotensin-aldosterone system activation and hepatic hepcidin suppression might indicate stress erythropoiesisinduced in HF. Furthermore, assembled ferritin increases but ferritin-bound Fe3+ is reduced in myocardium, indicating that a failing heart increases the iron storage reserve but iron deficiency leads to a drop in myocardial iron stores. Together, HF in pigs leads to down-regulated iron bioavailability and reduced hepatic iron storage making iron unavailable for systemic/cardiac needs.


Subject(s)
Heart Failure/metabolism , Hemosiderin/metabolism , Liver/metabolism , Tachycardia/complications , Animals , Disease Models, Animal , Ferritins/metabolism , Humans , Iron/metabolism , Male , Renin-Angiotensin System , Swine , Tachycardia/etiology , Tachycardia/metabolism
6.
Acta Derm Venereol ; 101(9): adv00558, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34515804

ABSTRACT

Current understanding of the underlying pathophysiology of hidradenitis suppurativa (HS) links the disease with proinflammatory activation and autoimmune processes. This study investigated serum levels of interleukin (IL)-22, a cytokine critically involved in epithelial homeostasis, in the context of the broad clinical spectrum of patients with HS. The study also assessed the relationship between serum IL-22 and pro-inflammatory activation (as evidenced by serum level of IL-6) and serum hepcidin (central regulator of systemic iron homeostasis). Serum concentrations of IL-22 were assessed in 74 patients with HS and 15 healthy subjects. Compared with healthy controls, patients with HS demonstrated decreased levels of serum IL-22 (median; interquartile range (IQR): 12.4 pg/ml (9.8; 23.5) vs 34.8 pg/ml (24.8; 39.8), p < 0.001 vs controls). Disease severity (assessed both with Hurley staging and Hidradenitis Suppurativa Severity Index) did not differentiate IL-22 levels (p > 0.1 in both comparisons). Serum levels of IL-22 and IL-6 did not correlate with each other (R=-0.17, p = ns). In a subgroup of 24 patients with HS with pro-inflammatory activation, the mean level of IL-22 was similar to that of the remaining patients (median (IQR): 9.8 pg/ml (8.5; 15.0) vs 12.0 pg/ml (9.4; 16.3), p = ns). Patients with HS demonstrated a decreased level of hepcidin (mean: 31.3 ± 25.9 pg/ml), which correlated with the levels of IL-22 (R=0.36, p <0.05). Patients with HS demonstrated significantly decreased levels of serum IL-22, which was neither correlated with pro-inflammatory status nor associated with disease severity, but correlated modestly with serum hepcidin.


Subject(s)
Hidradenitis Suppurativa , Cytokines , Hepcidins , Hidradenitis Suppurativa/diagnosis , Humans , Interleukins , Interleukin-22
7.
Cells ; 10(4)2021 04 06.
Article in English | MEDLINE | ID: mdl-33917391

ABSTRACT

Cardiac fibroblasts and cardiomyocytes are the main cells involved in the pathophysiology of myocarditis (MCD). These cells are especially sensitive to changes in iron homeostasis, which is extremely important for the optimal maintenance of crucial cellular processes. However, the exact role of iron status in the pathophysiology of MCD remains unknown. We cultured primary human cardiomyocytes (hCM) and cardiofibroblasts (hCF) with sera from acute MCD patients and healthy controls to mimic the effects of systemic inflammation on these cells. Next, we performed an initial small-scale (n = 3 per group) RNA sequencing experiment to investigate the global cellular response to the exposure on sera. In both cell lines, transcriptomic data analysis revealed many alterations in gene expression, which are related to disturbed canonical pathways and the progression of cardiac diseases. Moreover, hCM exhibited changes in the iron homeostasis pathway. To further investigate these alterations in sera-treated cells, we performed a larger-scale (n = 10 for controls, n = 18 for MCD) follow-up study and evaluated the expression of genes involved in iron metabolism. In both cell lines, we demonstrated an increased expression of transferrin receptor 1 (TFR1) and ferritin in MCD serum-treated cells as compared to controls, suggesting increased iron demand. Furthermore, we related TFR1 expression with the clinical profile of patients and showed that greater iron demand in sera-treated cells was associated with higher inflammation score (interleukin 6 (IL-6), C-reactive protein (CRP)) and advanced neurohormonal activation (NT-proBNP) in patients. Collectively, our data suggest that the malfunctioning of cardiomyocytes and cardiofibroblasts in the course of MCD might be related to alterations in the iron homeostasis.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Iron/metabolism , Myocarditis/blood , Myocytes, Cardiac/metabolism , Acute Disease , Adult , Case-Control Studies , Cell Survival , Cells, Cultured , Down-Regulation/genetics , Female , Ferritins/blood , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/pathology , Male , Middle Aged , Myocarditis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Treatment Outcome , Up-Regulation/genetics
8.
Int J Cardiol ; 322: 191-196, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32841617

ABSTRACT

BACKGROUND: Inflammatory responses play an important role in the pathophysiology of cardiogenic shock (CS). The aim of this study was to investigate the kinetics of procalcitonin (PCT), C-reactive protein (CRP), and interleukin-6 (IL-6) in CS and to assess their relation to clinical presentation, other biochemical variables, and prognosis. METHODS: Levels of PCT, CRP and IL-6 were analyzed in serial plasma samples (0-120h) from 183 patients in the CardShock study. The study population was dichotomized by PCTmax ≥ and < 0.5 µg/L, and IL-6 and CRPmax above/below median. RESULTS: PCT peaked already at 24 h [median PCTmax 0.71 µg/L (IQR 0.24-3.4)], whereas CRP peaked later between 48 and 72 h [median CRPmax 137 mg/L (59-247)]. PCT levels were significantly higher among non-survivors compared with survivors from 12 h on, as were CRP levels from 24 h on (p < 0.001). PCTmax ≥ 0.5 µg/L (60% of patients) was associated with clinical signs of systemic hypoperfusion, cardiac and renal dysfunction, acidosis, and higher levels of blood lactate, IL-6, growth-differentiation factor 15 (GDF-15), and CRPmax. Similarly, IL-6 > median was associated with clinical signs and biochemical findings of systemic hypoperfusion. PCTmax ≥ 0.5 µg/L and IL-6 > median were associated with increased 90-day mortality (50% vs. 30% and 57% vs. 22%, respectively; p < 0.01 for both), while CRPmax showed no prognostic significance. The association of inflammatory markers with clinical infections was modest. CONCLUSIONS: Inflammatory markers are highly related to signs of systemic hypoperfusion in CS. Moreover, high PCT and IL-6 levels are associated with poor prognosis.


Subject(s)
Interleukin-6 , Procalcitonin , Biomarkers , C-Reactive Protein/analysis , Humans , Kinetics , Prognosis , Shock, Cardiogenic/diagnosis
9.
J Vet Res ; 64(2): 325-332, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32587922

ABSTRACT

INTRODUCTION: Although peripheral blood analysis has become increasingly automated, microscopy is the only available method for the diagnosis of anisocytosis and poikilocytosis. The aims of the study were to compare RBC volume data obtained with two different analysers and by manual assessment of smears and to compare this data between dogs in various stages of heart failure secondary to degenerative mitral valvular (DMV) disease. The impact of diuretic administration on RBC morphology was also assessed. MATERIAL AND METHODS: Sixty-eight dogs, 56 in different stages of DMV disease and 12 as healthy controls, were studied. Impedance and flow cytometry haematological analyses were performed for each animal. Additionally, two smears were prepared for manual analysis. RBC structure, staining, and size differences were recorded. RESULTS: There were no significant differences between the blood morphological parameters assessed using haematological analysers nor between dogs receiving diuretic treatment and those not treated. Based on the manual smear, significantly higher erythrocyte anisocytosis was observed in the dogs with symptomatic DMV disease than in the control group. CONCLUSION: Haematological analysers based on impedance and flow cytometry provide reliable and comparable morphological results in dogs with heart failure. However, microscopic assessment of blood smears is a more reliable tool to detect erythrocyte anisocytosis.

10.
Dermatology ; 236(1): 52-58, 2020.
Article in English | MEDLINE | ID: mdl-31927542

ABSTRACT

BACKGROUND: Proinflammatory activation and autoimmune processes underlie the pathophysiology of hidradenitis suppurativa (HS). Iron deficiency (ID) is frequently present in inflammation-mediated chronic diseases, irrespective of anemia. OBJECTIVES: We aimed to characterize iron status in patients with HS. METHODS: Serum concentrations of ferritin, transferrin saturation (Tsat), soluble transferrin receptor and hepcidin were assessed as the biomarkers of iron status in 74 patients with HS and 44 healthy subjects. ID was defined as ferritin <100 µg/L or ferritin 100-299 µg/L with Tsat <20% (following the definition used in the other studies in chronic disease). RESULTS: Compared with controls, patients with HS demonstrated a deranged iron status as evidenced by decreased levels of ferritin (91 ± 87 vs. 157 ± 99 µg/L), Tsat (21.5 ± 10.8 vs. 42.2 ± 11.7%) and hepcidin (31.3 ± 25.9 vs. 44.2 ± 22.0 ng/mL) (all p < 0.05 vs. controls). There was also a trend toward higher values of soluble transferrin receptor (1.23 ± 0.35 vs. 1.12 ± 0.19 mg/L) (p = 0.09 vs. controls). Disease severity (assessed with the Hidradenitis Suppurativa Severity Index and the 3-degree Hurley scale) did not differentiate iron status biomarkers. ID was present in 75% of HS patients, and its prevalence was not related with disease severity (Hurley I/II/III - 82 vs. 73 vs. 67%). In HS, none of the iron status biomarkers correlated with the levels of interleukin-6 (a marker of proinflammatory activation). CONCLUSIONS: The majority of HS patients demonstrate derangements in iron status typical of ID. These abnormalities are neither related to proinflammatory activation nor associated with disease severity. Whether it may have a therapeutic impact needs to be further studied.


Subject(s)
Deficiency Diseases/blood , Hidradenitis Suppurativa/blood , Iron Deficiencies , Adult , Deficiency Diseases/complications , Deficiency Diseases/diagnosis , Female , Hidradenitis Suppurativa/complications , Humans , Iron/blood , Male , Middle Aged , Young Adult
11.
Heart Fail Rev ; 24(2): 269-277, 2019 03.
Article in English | MEDLINE | ID: mdl-30284070

ABSTRACT

Iron deficiency (ID) is a common and ominous comorbidity in heart failure (HF) and predicts worse outcomes, independently of the presence of anaemia. Accumulated data from animal models of systemic ID suggest that ID is associated with several functional and structural abnormalities of the heart. However, the exact role of myocardial iron deficiency irrespective of systemic ID and/or anaemia has been elusive. Recently, several transgenic models of cardiac-specific ID have been developed to investigate the influence of ID on cardiac tissue. In this review, we discuss structural and functional cardiac consequences of ID in these models and summarize data from clinical studies. Moreover, the beneficial effects of intravenous iron supplementation are specified.


Subject(s)
Anemia, Iron-Deficiency/complications , Heart Failure/physiopathology , Heart/physiopathology , Iron Deficiencies , Iron/blood , Administration, Intravenous , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Comorbidity , Female , Heart/drug effects , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/mortality , Hepcidins/metabolism , Homeostasis/physiology , Humans , Iron/administration & dosage , Iron/therapeutic use , Iron Metabolism Disorders/complications , Male , Mice , Mice, Transgenic/metabolism , Models, Animal , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, Transferrin/metabolism
12.
Bioconjug Chem ; 29(11): 3509-3515, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30365887

ABSTRACT

Anionic boron clusters are man-made, inorganic compounds with potential applications in therapeutic peptides modification to improve their biological activity and pharmacokinetics, e.g., by enabling complexation with serum albumin. However, the conjugation of anionic boron clusters and peptides remains poorly understood. Here, we report a solid-state, thermal reaction to selectively conjugate carboxylic groups in the peptide thymosin ß4 (Tß4) with cyclic oxonium derivatives of anionic boron clusters (dodecaborate anion [B12H12]2- and cobalt bis(1,2-dicarbollide), [COSAN]- [3,3'-Co(1,2-C2B9H11)2]-). Modification of the carboxylic groups retains the negative charge at the modification site and leads to the formation of ester bonds. The ester bonds in the conjugates undergo hydrolysis at different rates depending on the site of the modification. We obtained conjugates with dramatically different stabilities (τ1/2 from 3-836 h (Tß4-[B12H12]2- conjugates) and 9-1329 h (Tß4-[COSAN]- conjugates)) while retaining or improving the prosurvival activity of Tß4 toward cardiomyocytes (H9C2 cell line).


Subject(s)
Boron/chemistry , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Thymosin/chemistry , Anions/chemistry , Cell Line , Coordination Complexes/pharmacokinetics , Half-Life , Humans , Hydrogen-Ion Concentration , Hydrolysis , Myocytes, Cardiac/drug effects , Serum Albumin/chemistry
13.
Cells ; 7(10)2018 Oct 20.
Article in English | MEDLINE | ID: mdl-30347796

ABSTRACT

(1) Background: Oxidative energy metabolism is presumed to rely on the optimal iron supply. Primary human cardiac myocytes (HCM) exposed to different iron availability conditions during mechanical stretch are anticipated to demonstrate expression changes of genes involved in aerobic and anaerobic metabolic pathways. (2) Methods: HCM were cultured for 48 h either in static conditions and upon mechanical stretch at the optimal versus reduced versus increased iron concentrations. We analyzed the expression of pyruvate kinase (PKM2), lactate dehydrogenase A (LDHA), and mitochondrial complexes I⁻V at the mRNA and protein levels. The concentration of l-lactate was assessed by means of lactate oxidase method-based kit. (3) Results: Reduced iron concentrations during mechanical work caused a decreased expression of complexes I⁻V (all p < 0.05). The expression of PKM2 and LDHA, as well as the medium concentration of l-lactate, was increased in these conditions (both p < 0.05). HCM exposed to the increased iron concentration during mechanical effort demonstrated a decreased expression of mitochondrial complexes (all p < 0.01); however, a decrement was smaller than in case of iron chelation (p < 0.05). The iron-enriched medium caused a decrease in expression of LDHA and did not influence the concentration of l-lactate. (4) Conclusions: During mechanical effort, the reduced iron availability enhances anaerobic glycolysis and extracellular lactate production, whilst decreasing mitochondrial aerobic pathway in HCM. Iron enrichment during mechanical effort may be protective in the context of intracellular protein machinery of non-oxidative metabolism with no effect on the extracellular lactate concentration.

14.
J Cachexia Sarcopenia Muscle ; 9(5): 802-815, 2018 10.
Article in English | MEDLINE | ID: mdl-30178922

ABSTRACT

Specific skeletal myopathy constitutes a common feature of heart failure, chronic obstructive pulmonary disease, and type 2 diabetes mellitus, where it can be characterized by the loss of skeletal muscle oxidative capacity. There is evidence from in vitro and animal studies that iron deficiency affects skeletal muscle functioning mainly in the context of its energetics by limiting oxidative metabolism in favour of glycolysis and by alterations in both carbohydrate and fat catabolic processing. In this review, we depict the possible molecular pathomechanisms of skeletal muscle energetic impairment and postulate iron deficiency as an important factor causally linked to loss of muscle oxidative capacity that contributes to skeletal myopathy seen in patients with heart failure, chronic obstructive pulmonary disease, and type 2 diabetes mellitus.


Subject(s)
Disease Susceptibility , Energy Metabolism , Iron Deficiencies , Muscle, Skeletal/metabolism , Stress, Physiological , Animals , Chronic Disease , Humans , Muscular Diseases/etiology , Muscular Diseases/metabolism , Muscular Diseases/therapy , Oxidation-Reduction , Oxygen/metabolism
15.
Biomed Res Int ; 2018: 6982897, 2018.
Article in English | MEDLINE | ID: mdl-29721510

ABSTRACT

BACKGROUND: Acute heart failure (AHF) is associated with disturbances of the peripheral perfusion leading to the dysfunction of many organs. Consequently, an episode of AHF constitutes a "multiple organ failure" which may also affect the skeletal muscles. However, the abnormalities within skeletal muscles during AHF have not been investigated so far. The aim of this project is to comprehensively evaluate skeletal muscles (at a functional and tissue level) during AHF. METHODS: The study will include ≥63 consecutive AHF patients who will be randomized into 2 groups: ≥42 with cardiac rehabilitation group versus ≥21 with standard pharmacotherapy alone. The following tests will be conducted on the first and last day of hospitalization, at rest and after exercise, and 30 days following the discharge: clinical evaluation, medical interview, routine physical examination, echocardiography, and laboratory tests (including the assessment of NT-proBNP, inflammatory markers, and parameters reflecting the status of the kidneys and the liver); hemodynamic evaluation, noninvasive determination of cardiac output and systemic vascular resistance using the impedance cardiography; evaluation of biomarkers reflecting myocyte damage, immunochemical measurements of tissue-specific enzymatic isoforms; evaluation of skeletal muscle function, using surface electromyography (sEMG) (maximum tonus of the muscles will be determined along with the level of muscular fatigability); evaluation of muscle tissue perfusion, assessed on the basis of the oxygenation level, with noninvasive direct continuous recording of perfusion in peripheral tissues by local tissue oximetry, measured by near-infrared spectroscopy (NIRS). RESULTS AND CONCLUSIONS: Our findings will demonstrate that the muscle tissue is another area of the body which should be taken into consideration in the course of treatment of AHF, requiring a development of targeted therapeutic strategies, such as a properly conducted rehabilitation.


Subject(s)
Heart Failure/physiopathology , Heart Failure/rehabilitation , Muscle, Skeletal/physiopathology , Acute Disease , Female , Humans , Male
16.
Int J Mol Med ; 41(5): 2678-2686, 2018 May.
Article in English | MEDLINE | ID: mdl-29436580

ABSTRACT

There is clinical evidence that patients with heart failure and concomitant iron deficiency have increased skeletal muscle fatigability and impaired exercise tolerance. It was expected that a skeletal muscle cell line subjected to different degrees of iron availability and/or concomitant hypoxia would demonstrate changes in cell morphology and in the expression of atrophy markers. L6G8C5 rat skeletal myocytes were cultured in normoxia or hypoxia at optimal, reduced or increased iron concentrations. Experiments were performed to evaluate the iron content in cells, cell morphology, and the expression of muscle specific atrophy markers [Atrogin1 and muscle­specific RING­finger 1 (MuRF1)], a gene associated with the atrophy/hypertrophy balance [mothers against decapentaplegic homolog 4 (SMAD4)] and a muscle class­III intermediate filament protein (Desmin) at the mRNA and protein level. Hypoxic treatment caused, as compared to normoxic conditions, an increase in the expression of Atrogin­1 (P<0.001). Iron­deficient cells exhibited morphological abnormalities and demonstrated a significant increase in the expression of Atrogin­1 (P<0.05) and MuRF1 (P<0.05) both in normoxia and hypoxia, which indicated activation of the ubiquitin proteasome pathway associated with protein degradation during muscle atrophy. Depleted iron in cell culture combined with hypoxia also induced a decrease in SMAD4 expression (P<0.001) suggesting modifications leading to atrophy. In contrast, cells cultured in a medium enriched with iron during hypoxia exhibited inverse changes in the expression of atrophy markers (both P<0.05). Desmin was upregulated in cells subjected to both iron depletion and iron excess in normoxia and hypoxia (all P<0.05), but the greatest augmentation of mRNA expression occurred when iron depletion was combined with hypoxia. Notably, in hypoxia, an increased expression of Atrogin­1 and MuRF1 was associated with an increased expression of transferrin receptor 1, reflecting intracellular iron demand (R=0.76, P<0.01; R=0.86, P<0.01). Hypoxia and iron deficiency when combined exhibited the most detrimental impact on skeletal myocytes, especially in the context of muscle atrophy markers. Conversely, iron supplementation in in vitro conditions acted in a protective manner on these cells.


Subject(s)
Deferoxamine/pharmacology , Ferric Compounds/pharmacology , Iron/metabolism , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy/metabolism , Muscular Atrophy/prevention & control , Quaternary Ammonium Compounds/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Line , Desmin/genetics , Gene Expression Regulation/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Rats , Smad4 Protein/genetics
17.
Kardiol Pol ; 75(3): 267-275, 2017.
Article in English | MEDLINE | ID: mdl-27747853

ABSTRACT

BACKGROUND: Iron is presumed to play an important role in the functioning of cardiomyocytes and skeletal myocytes. There is scarcity of direct data characterising the cells functioning when exposed to iron depletion or iron overload in a cellular environment. There is some clinical evidence demonstrating that iron deficiency has serious negative prognostic consequences in heart failure (HF) patients and its correction brought clinical benefit. AIM: The viability of the cells upon unfavourable iron concentration in the cell culture medium and the presence of the molecular system of proteins involved in intracellular iron metabolism in these cells have been studied. METHODS: H9C2 rat adult cardiomyocytes and L6G8C5 rat adult skeletal myocytes were cultured for 24 h in optimal vs. reduced vs. increased iron concentrations. Intracellular iron content was measured by flame atomic absorption spectroscopy (FAAS). We analysed the mRNA expression of: ferritin heavy and light chains (FTH and FTL; iron storage proteins), myoglobin (MB, oxygen storage protein) ferroportin type 1 (FPN1; iron exporter), transferrin receptor type 1 (TfR1; iron importer), hepcidin (HAMP; iron metabolism regulator) using qPCR, the level of respective proteins using Western Blot (WB), and the viability of the cells using flow cytometry and cell viability tetrazolium reduction assay (MTS). RESULTS: Cardiomyocytes exposed to gradually reduced iron concentrations in the medium demonstrated a decrease in the mRNA expression of FTH, FTL, FPN1, MB, and HAMP (all R = -0.75, p < 0.05), indicating depleted iron status in the cells. As a consequence, the expression of TfR1 (R = 0.7, p < 0.05) was increased, reflecting a facilitated entrance of iron to the cells. The inverse changes occurred in H9C2 cells exposed to increased iron concentrations in the medium in comparison to control cells. The same pattern of changes in the mRNA expressions was observed in myocytes, and there was a strong correlation between analogous genes in both cell lines (all R > 0.9, p < 0.0001). WB analysis revealed the analogous pattern of changes in protein expression as an mRNA profile. Both iron depletion and iron excess impaired viability of cardiomyocytes and skeletal myocytes. CONCLUSIONS: Both rat cardiomyocytes and myocyte cells contain the set of genes involved in the intracellular iron metabolism, and both types of investigated cells respond to changing iron concentrations in the cultured environment. Both iron deficiency (ID) and iron overload is detrimental for the cells. This data may explain the beneficial effects of iron supplementation in patients with ID in HF.


Subject(s)
Iron/physiology , Muscle Cells/physiology , Nutritional Status , Anemia, Iron-Deficiency , Animals , Antigens, CD/genetics , Cation Transport Proteins/genetics , Cell Line , Cell Survival , Ferritins/genetics , Gene Expression Regulation , Hepcidins/genetics , Iron/analysis , Iron/metabolism , Iron Overload , Muscle Cells/metabolism , Rats , Receptors, Transferrin/genetics
18.
Mol Med Rep ; 14(4): 3969-77, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27599775

ABSTRACT

The differential availability of iron during hypoxia is presumed to affect the functioning of cardiac and skeletal myocytes. Rat H9C2 cardiomyocytes and L6G8C5 myocytes were cultured for 48 h in normoxic or hypoxic conditions at the optimal, reduced or increased iron concentration. The mRNA expression levels of markers of apoptosis [B­cell lymphoma­2 (Bcl2; inhibition) and Bcl­2­activated X protein (Bax; induction)], atrophy (Atrogin), glycolysis (pyruvate kinase 2; PKM2) and iron metabolism [transferrin receptor 1 (TfR1; iron importer), ferroportin 1 (FPN1; iron exporter), ferritin heavy chain (FTH; iron storage protein) and hepcidin (HAMP; iron regulator)] were determined using reverse transcription­quantitative polymerase chain reaction, and cell viability was measured using an tetrazolium reduction assay. Cardiomyocytes and myocytes, when exposed to hypoxia, demonstrated an increased Bax/Bcl­2 gene expression ratio (P<0.05). Additional deferoxamine (DFO) treatment resulted in further increases in Bax/Bcl­2 in each cell type (P<0.001 each) and this was associated with the 15% loss in viability. The analogous alterations were observed in both cell types upon ammonium ferric citrate (AFC) treatment during hypoxia; however, the increased Bax/Bcl­2 ratio and associated viability loss was lower compared with that in case of DFO treatment (P<0.05 each). Under hypoxic conditions, myocytes demonstrated an increased expression of PKM2 (P<0.01). Additional DFO treatment caused an increase in the mRNA expression levels of PKM2 and Atrogin­1 (P<0.001 and P<0.05, respectively), whereas AFC treatment caused an increased mRNA expression of PKM2 (P<0.01) and accompanied decreased mRNA expression of Atrogin­1 (P<0.05). The expression augmentation of PKM2 during hypoxia was greater upon low iron compared with that of ferric salt treatment (P<0.01). Both cell types upon DFO during hypoxia demonstrated the increased expression of TfR1 and HAMP (all P<0.05), which was associated with the increased Bax/Bcl­2 ratio (all R>0.6 and P<0.05). In conclusion, during hypoxia iron deficiency impairs the viability of cardiomyocytes and myocytes more severely compared with iron excess. In myocytes, during hypoxia iron may act in a protective manner, since the level of atrophy is decreased in the iron­salt­treated cells.


Subject(s)
Apoptosis , Gene Expression Regulation , Iron/metabolism , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Hypoxia , Cell Line , Muscle Fibers, Skeletal/cytology , Myocytes, Cardiac/cytology , Oxygen/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , bcl-2-Associated X Protein/genetics
20.
Eur J Heart Fail ; 18(7): 762-73, 2016 07.
Article in English | MEDLINE | ID: mdl-26800032

ABSTRACT

Skeletal and respiratory myopathy not only constitutes an important pathophysiological feature of heart failure and chronic obstructive pulmonary disease, but also contributes to debilitating symptomatology and predicts worse outcomes in these patients. Accumulated evidence from laboratory experiments, animal models, and interventional studies in sports medicine suggests that undisturbed systemic iron homeostasis significantly contributes to the effective functioning of skeletal muscles. In this review, we discuss the role of iron status for the functioning of skeletal muscle tissue, and highlight iron deficiency as an emerging therapeutic target in chronic diseases accompanied by a marked muscle dysfunction.


Subject(s)
Anemia, Iron-Deficiency/physiopathology , Deficiency Diseases/physiopathology , Iron Deficiencies , Muscle, Skeletal/physiopathology , Anemia, Iron-Deficiency/metabolism , Animals , Chronic Disease , Deficiency Diseases/metabolism , Energy Metabolism , Exercise Tolerance , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , In Vitro Techniques , Iron/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Muscles/metabolism , Respiratory Muscles/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...