Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.083
Filter
1.
Nat Commun ; 15(1): 8131, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39284836

ABSTRACT

Hematopoietic stem cells (HSCs) react to various stress conditions. However, it is unclear whether and how HSCs respond to severe anemia. Here, we demonstrate that upon induction of acute anemia, HSCs rapidly proliferate and enhance their erythroid differentiation potential. In severe anemia, lipoprotein profiles largely change and the concentration of ApoE increases. In HSCs, transcription levels of lipid metabolism-related genes, such as very low-density lipoprotein receptor (Vldlr), are upregulated. Stimulation of HSCs with ApoE enhances their erythroid potential, whereas HSCs in Apoe knockout mice do not respond to anemia induction. VldlrhighHSCs show higher erythroid potential, which is enhanced after acute anemia induction. VldlrhighHSCs are epigenetically distinct because of their low chromatin accessibility, and more chromatin regions are closed upon acute anemia induction. Chromatin regions closed upon acute anemia induction are mainly binding sites of Erg. Inhibition of Erg enhanced the erythroid differentiation potential of HSCs. Our findings indicate that lipoprotein metabolism plays an important role in HSC regulation under severe anemic conditions.


Subject(s)
Anemia , Apolipoproteins E , Cell Differentiation , Hematopoietic Stem Cells , Lipoproteins , Animals , Anemia/metabolism , Anemia/genetics , Hematopoietic Stem Cells/metabolism , Mice , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Lipoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/metabolism , Receptors, LDL/genetics , Male , Chromatin/metabolism , Erythropoiesis/genetics , Erythroid Cells/metabolism
2.
J Agric Food Chem ; 72(32): 18155-18161, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39088813

ABSTRACT

Balkan endemic nephropathy (BEN) is a chronic kidney disease that predominantly affects inhabitants of rural farming communities along the Danube River tributaries in the Balkans. Long-standing research has identified dietary exposure to aristolochic acids (AAs) as the principal toxicological cause. This study investigates the pathophysiological role of anemia in BEN, noting its earlier and more severe manifestation in BEN patients compared to those with other chronic kidney diseases. Utilizing a mouse model, our research demonstrates that prolonged exposure to aristolochic acid I (AA-I) (the most prevalent AA variant) leads to significant red blood cell depletion through DNA damage, such as DNA adduct formation in bone marrow, prior to observable kidney function decline. Furthermore, in vitro experiments with kidney cells exposed to lowered oxygen and pH conditions mimicking an anemia environment show enhanced DNA adduct formation, suggesting increased AA-I mutagenicity and carcinogenicity. These findings indicate for the first time a positive feedback mechanism of AA-induced anemia, DNA damage, and kidney impairment in BEN progression. These results not only advance our understanding of the underlying mechanisms of BEN but also highlight anemia as a potential target for early BEN diagnosis and therapy.


Subject(s)
Anemia , Aristolochic Acids , Balkan Nephropathy , DNA Adducts , Aristolochic Acids/toxicity , Aristolochic Acids/adverse effects , Balkan Nephropathy/chemically induced , Balkan Nephropathy/metabolism , Balkan Nephropathy/genetics , DNA Adducts/metabolism , Animals , Mice , Humans , Anemia/chemically induced , Anemia/metabolism , Anemia/genetics , Male , DNA Damage/drug effects , Mice, Inbred C57BL , Kidney/drug effects , Kidney/metabolism , Female
3.
Nat Commun ; 15(1): 5678, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971858

ABSTRACT

Inherited non-hemolytic anemia is a group of rare bone marrow disorders characterized by erythroid defects. Although concerted efforts have been made to explore the underlying pathogenetic mechanisms of these diseases, the understanding of the causative mutations are still incomplete. Here we identify in a diseased pedigree that a gain-of-function mutation in toll-like receptor 8 (TLR8) is implicated in inherited non-hemolytic anemia. TLR8 is expressed in erythroid lineage and erythropoiesis is impaired by TLR8 activation whereas enhanced by TLR8 inhibition from erythroid progenitor stage. Mechanistically, TLR8 activation blocks annexin A2 (ANXA2)-mediated plasma membrane localization of STAT5 and disrupts EPO signaling in HuDEP2 cells. TLR8 inhibition improves erythropoiesis in RPS19+/- HuDEP2 cells and CD34+ cells from healthy donors and inherited non-hemolytic anemic patients. Collectively, we identify a gene implicated in inherited anemia and a previously undescribed role for TLR8 in erythropoiesis, which could potentially be explored for therapeutic benefit in inherited anemia.


Subject(s)
Anemia , Erythropoiesis , Toll-Like Receptor 8 , Humans , Erythropoiesis/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , Female , Anemia/genetics , Male , Pedigree , Erythropoietin/metabolism , Erythropoietin/genetics , Adult , Signal Transduction , Mutation , Erythroid Cells/metabolism , Animals , Erythroid Precursor Cells/metabolism
4.
Nat Commun ; 15(1): 5037, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866743

ABSTRACT

This study on severe malarial anemia (SMA: Hb < 6.0 g/dL), a leading global cause of childhood morbidity and mortality, compares the entire expressed whole blood host transcriptome between Kenyan children (3-48 mos.) with non-SMA (Hb ≥ 6.0 g/dL, n = 39) and SMA (n = 18). Differential expression analyses reveal 1403 up-regulated and 279 down-regulated transcripts in SMA, signifying impairments in host inflammasome activation, cell death, and innate immune and cellular stress responses. Immune cell profiling shows decreased memory responses, antigen presentation, and immediate pathogen clearance, suggesting an immature/improperly regulated immune response in SMA. Module repertoire analysis of blood-specific gene signatures identifies up-regulation of erythroid genes, enhanced neutrophil activation, and impaired inflammatory responses in SMA. Enrichment analyses converge on disruptions in cellular homeostasis and regulatory pathways for the ubiquitin-proteasome system, autophagy, and heme metabolism. Pathway analyses highlight activation in response to hypoxic conditions [Hypoxia Inducible Factor (HIF)-1 target and Reactive Oxygen Species (ROS) signaling] as a central theme in SMA. These signaling pathways are also top-ranking in protein abundance measures and a Ugandan SMA cohort with available transcriptomic data. Targeted RNA-Seq validation shows strong concordance with our entire expressed transcriptome data. These findings identify key molecular themes in SMA pathogenesis, offering potential targets for new malaria therapies.


Subject(s)
Anemia , Transcriptome , Humans , Anemia/genetics , Anemia/blood , Child, Preschool , Infant , Female , Malaria/blood , Malaria/genetics , Kenya , Male , Gene Expression Profiling , Immunity, Innate/genetics , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/blood
5.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38892362

ABSTRACT

During the first era of humanity, the conditions of life, including hunting, fighting, obtaining food, and diseases, were associated with frequent hemorrhages, anemia, and infections, which led to death or untreatable conditions [...].


Subject(s)
Erythrocytes , Humans , Erythrocytes/metabolism , Animals , Anemia/genetics
6.
Nat Commun ; 15(1): 5090, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918373

ABSTRACT

The development of haematopoiesis involves the coordinated action of numerous genes, some of which are implicated in haematological malignancies. However, the biological function of many genes remains elusive and unknown functional genes are likely to remain to be uncovered. Here, we report a previously uncharacterised gene in haematopoiesis, identified by screening mutant embryonic stem cells. The gene, 'attenuated haematopoietic development (Ahed)', encodes a nuclear protein. Conditional knockout (cKO) of Ahed results in anaemia from embryonic day 14.5 onward, leading to prenatal demise. Transplantation experiments demonstrate the incapacity of Ahed-deficient haematopoietic cells to reconstitute haematopoiesis in vivo. Employing a tamoxifen-inducible cKO model, we further reveal that Ahed deletion impairs the intrinsic capacity of haematopoietic cells in adult mice. Ahed deletion affects various pathways, and published databases present cancer patients with somatic mutations in Ahed. Collectively, our findings underscore the fundamental roles of Ahed in lifelong haematopoiesis, implicating its association with malignancies.


Subject(s)
Hematopoiesis , Mice, Knockout , Animals , Hematopoiesis/genetics , Mice , Humans , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice, Inbred C57BL , Mutation , Anemia/genetics , Male , Embryonic Stem Cells/metabolism
7.
Clin Immunol ; 265: 110290, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944365

ABSTRACT

OBJECTIVE: Juvenile arthritis caused by loss-of-function LACC1 mutations is characterized by early onset of symmetric and chronic arthritis, associated with an elevation of inflammatory markers. We aimed to describe serum cytokine levels, explore the type I interferon pathway, and evaluate the efficacy of treatment in a patient presenting with polyarthritis and anemia caused by novel compound heterozygous variations in LACC1. METHODS: Clinical data of a patient with compound heterozygous variations in LACC1 was collected. Serum cytokine levels and IFN-stimulated cytokine genes were analyzed at diagnosis, at disease flare, and after treatment. Full-length cDNA of LACC1 was checked by RNA analysis. Single-cell RNA sequencing was performed in PBMCs. RESULTS: Two novel variants in the LACC1 gene were identified in a patient presenting with polyarthritis and anemia. LACC1-cDNA was normally expressed in the healthy control, the target production at 1384 bp was not observed in the patient. Compared to nine patient controls with non-systemic juvenile idiopathic arthritis, serum interleukin(IL)-6 level was significantly elevated in the affected patient. The median IFN score for the patient, her mother, and controls were 118, 8, and 4.9, respectively. The combined treatment of JAK inhibitors with prednisone or tocilizumab led to a complete response, including remission of joint symptoms, resolution of anemia, reduced expression of IFN-stimulated cytokine genes, and normalized levels of inflammatory markers, including CRP, ESR, SAA, and serum IL-6. CONCLUSION: LACC1 may play a crucial role in multiple inflammatory signaling pathways. The combination therapy of JAK inhibitors and tocilizumab may be effective for a subset of refractory patients.


Subject(s)
Anemia , Arthritis, Juvenile , Humans , Anemia/genetics , Anemia/drug therapy , Anemia/etiology , Female , Arthritis, Juvenile/genetics , Arthritis, Juvenile/drug therapy , Arthritis, Juvenile/complications , Arthritis, Juvenile/blood , Antibodies, Monoclonal, Humanized/therapeutic use , Prednisone/therapeutic use , Child , Cytokines/blood , Janus Kinase Inhibitors/therapeutic use , Interleukin-6/blood , Interleukin-6/genetics , Mutation , Intracellular Signaling Peptides and Proteins
8.
Free Radic Biol Med ; 219: 153-162, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657753

ABSTRACT

The anemia of inflammation (AI) is characterized by the presence of inflammation and abnormal elevation of hepcidin. Accumulating evidence has proved that Rocaglamide (RocA) was involved in inflammation regulation. Nevertheless, the role of RocA in AI, especially in iron metabolism, has not been investigated, and its underlying mechanism remains elusive. Here, we demonstrated that RocA dramatically suppressed the elevation of hepcidin and ferritin in LPS-treated mice cell line RAW264.7 and peritoneal macrophages. In vivo study showed that RocA can restrain the depletion of serum iron (SI) and transferrin (Tf) saturation caused by LPS. Further investigation showed that RocA suppressed the upregulation of hepcidin mRNA and downregulation of Fpn1 protein expression in the spleen and liver of LPS-treated mice. Mechanistically, this effect was attributed to RocA's ability to inhibit the IL-6/STAT3 pathway, resulting in the suppression of hepcidin mRNA and subsequent increase in Fpn1 and TfR1 expression in LPS-treated macrophages. Moreover, RocA inhibited the elevation of the cellular labile iron pool (LIP) and reactive oxygen species (ROS) induced by LPS in RAW264.7 cells. These findings reveal a pivotal mechanism underlying the roles of RocA in modulating iron homeostasis and also provide a candidate natural product on alleviating AI.


Subject(s)
Benzofurans , Hepcidins , Homeostasis , Interleukin-6 , Iron , Animals , Mice , Anemia/metabolism , Anemia/genetics , Anemia/drug therapy , Anemia/pathology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Ferritins/metabolism , Ferritins/genetics , Gene Expression Regulation/drug effects , Hepcidins/drug effects , Hepcidins/genetics , Hepcidins/metabolism , Homeostasis/drug effects , Inflammation/metabolism , Inflammation/genetics , Inflammation/pathology , Interleukin-6/metabolism , Interleukin-6/genetics , Iron/metabolism , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Macrophages/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Benzofurans/pharmacology
9.
Cancer Chemother Pharmacol ; 94(1): 103-108, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38652271

ABSTRACT

OBJECTIVE: BRCA1/2 are integral to the DNA repair mechanism and their germline pathogenic variants (gBRCA) result in a high risk for developing breast and ovarian cancer. Patients with gBRCA mutations showed increased sensitivity to DNA cross-linking agent but might have increased treatment-related toxicities. Thus, we hypothesized that gBRCA mutation ovarian cancer patients who underwent platinum-based chemotherapy might be at higher risk of developing chemotherapy-induced hematologic toxicity. METHODS: This study enrolled 160 patients with ovarian cancer who received frontline platinum-based chemotherapy between 2011 and 2019 in Kyungpook National University Chilgok Hospital. Incidence rate and severity of chemotherapy-induced hematologic toxicity (neutropenia, anemia, thrombocytopenia) was compared for BRCA mutation and wild patients. RESULTS: 160 women, including 62 BRCA1/2 (38 BRCA1, and 25 BRCA2) mutation group, and 98 noncarriers, were analyzed. A higher frequency of G2 anemia was noted in the BRCA -mutant group (22% vs. 1%, p = 0.07). Furthermore, G3 anemia was significantly common among BRCA group (12.9% vs. 3%, p = 0.02). In the subgroup analysis according to BRCA1/2 status, BRCA1 mutated patients showed a significantly higher frequency of G1 anemia than BRCA2 (89% vs. 60%, p = 0.01). In terms of neutropenia and thrombocytopenia, BRCA mutated patients and noncarriers had similar hematologic toxicity. CONCLUSION: Germline BRCA mutations were associated with a higher frequency of G2/3 anemia in ovarian cancer patients who underwent first-line platinum-based chemotherapy. Moreover, the BRCA1 mutation appeared to be more strongly associated with the incidence of chemotherapy-induced anemia. Our findings warrant further investigation in larger, prospective studies to confirm these current findings and determine whether preventive interventions may be necessary.


Subject(s)
BRCA1 Protein , BRCA2 Protein , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Middle Aged , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Aged , Adult , Germ-Line Mutation , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Anemia/chemically induced , Anemia/genetics , Retrospective Studies , Thrombocytopenia/chemically induced , Thrombocytopenia/epidemiology , Thrombocytopenia/genetics , Hematologic Diseases/chemically induced , Hematologic Diseases/epidemiology , Mutation
10.
Aging Male ; 27(1): 2346312, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38685728

ABSTRACT

BACKGROUND: Previous research has shown that testosterone deficiency (TD) increases the risk of anemia, but it is unclear whether anemia affects testosterone levels. This study investigated the influence of anemia on testosterone levels. METHODS: Utilizing data from six NHANES cycles, including demographic, testosterone levels, and hemoglobin concentrations, we employed multivariable-adjusted logistic regression to investigate the relationship between anemia and testosterone levels. Moreover, a two-sample Mendelian randomization (MR) study employing genome-wide association study (GWAS) data examined the causal relationship. Kaplan-Meier survival estimation was used to compared the overall survival (OS) of anemic and nonanemic patients with low testosterone and normal testosterone levels. RESULTS: The inclusion of 21,786 participants (2318 with anemia and19,468 without anemia) revealed that nonanemic patients exhibited higher testosterone levels than did anemic patients (ß = 22.616, 95% CI: 3.873-41.359, p = 0.01807). MR analysis confirmed anemia as a cause of TD (OR = 1.045, 95% CI: 1.020-1.071, p < 0.001). Anemic males with low testosterone had reduced OS compared to those with normal levels (p < 0.001). CONCLUSIONS: Anemia emerged as a potential risk factor for TD, highlighting a bidirectional relationship between these conditions. Additional prospective investigations are essential for the validation and reinforcement of our findings.


Subject(s)
Anemia , Genome-Wide Association Study , Mendelian Randomization Analysis , Nutrition Surveys , Testosterone , Humans , Testosterone/blood , Testosterone/deficiency , Male , Anemia/genetics , Anemia/epidemiology , Middle Aged , Adult , Aged , Risk Factors
12.
BMC Infect Dis ; 24(1): 301, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38468199

ABSTRACT

BACKGROUND: Globally, 80 million people are suffering from chronic Hepatitis C virus (HCV) infection. Sofosbuvir ribavirin-based anti-HCV therapy is associated with anemia and other adverse effects. Polymorphisms of Inosine triphosphatase (ITPA) gene may cause functional impairment in the Inosine triphosphate pyrophosphatase enzyme, resulting in enhanced sustained viral response (SVR) and protection from ribavirin-associated anemia in patients on therapy. The study objective was to investigate the effect of Inosine triphosphatase gene polymorphism on SVR achievement, hemoglobin decline and ribavirin dose reduction in patients on therapy. METHODS: This prospective cohort study was of 170 hepatitis C infected patients received 6-month sofosbuvir ribavirin therapy. Patient viral load, reduction in ribavirin amount, liver function test, and complete blood count were noted monthly. Inosine triphosphatase variants rs1127354 and rs7270101 were assessed through the restriction fragment length polymorphism and confirmed using Sanger sequencing. The impact of polymorphism on cumulative reduction of ribavirin, and anti-HCV therapy outcome were studied. RESULTS: A total of 74.3% of patients had ITPA rs1127354 CC genotype, 25.7% were CA and AA 0%. The frequency of ITPA genotype rs7270101-AA was 95%, AC 5%, and CC was 0%. ITPA rs1127354-CA had a notably positive impact on SVR achievement with a zero-relapse rate. ITPA rs1127354-CA genotype was significantly (P ˂0.05) protective against ≥ 2 g/dl Hb reduction from baseline to 1st, 2nd and 6th months of therapy. During treatment, Hb reduction ≥ 10 g/dl was frequently observed in rs1127354-CC genotype and rs7270101-AA genotype patients. Ribavirin dose reduction was significantly (P ˂0.05) high in rs1127354-CC genotype as compared to genotype CA whereas no significant difference was observed in ribavirin dose reduction in rs7270101 AA and non-AA genotype. Patient baseline characteristics such as age, body mass index, rs1127354-CC genotype, and baseline Hb were significantly associated with significant Hb reduction. CONCLUSION: Pretreatment evaluation of ITPA polymorphism can be a diagnostic tool to find out patients at risk of anemia and improve treatment adherence. ITPA genotype rs1127354-CA contributes to improved compliance with ribavirin dose and protects against hemoglobin decline in HCV patients while taking ribavirin-based therapy. However, ITPA rs1127354, rs7270101 polymorphism have no significant impact on SVR achievement.


Subject(s)
Anemia , Hepatitis C, Chronic , Hepatitis C , Humans , Ribavirin/adverse effects , Sofosbuvir/adverse effects , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Antiviral Agents/adverse effects , Inosine Triphosphatase , Hepacivirus/genetics , Prospective Studies , Polymorphism, Single Nucleotide , Pyrophosphatases/genetics , Pyrophosphatases/therapeutic use , Anemia/chemically induced , Anemia/genetics , Hepatitis C/drug therapy , Genotype , Hemoglobins/genetics , Treatment Outcome
13.
Exp Hematol ; 132: 104174, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38331018

ABSTRACT

Hematopoietic stem cell (HSC) transplantation is extensively studied in mouse models, but their limited scale presents challenges for effective engraftment and comprehensive evaluations. Rats, owing to their larger size and anatomical similarity to humans, offer a promising alternative. In this study, we establish a rat model with the KitV834M mutation, mirroring KitW41 mice often used in KIT signaling and HSC research. KitV834M rats are viable and fertile, displaying anemia and mast cell depletion similar to KitW41 mice. The colony-forming unit assay revealed that the KitV834M mutation leads to reduced proliferation and loss of or decreased pluripotency of hematopoietic stem and progenitor cells (HSPCs), resulting in diminished competitive repopulating capacity of KitV834M HSPCs in competitive transplantation assays. Importantly, KitV834M rats support donor rat-HSC engraftment without irradiation. Leveraging the larger scale of this rat model enhances our understanding of HSC biology and transplantation dynamics, potentially advancing our knowledge in this field.


Subject(s)
Anemia , Hematopoietic Stem Cell Transplantation , Humans , Mice , Animals , Rats , Hematopoietic Stem Cells , Colony-Forming Units Assay , Anemia/genetics , Mutation , Mice, Inbred C57BL
14.
Haematologica ; 109(9): 2955-2968, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38385251

ABSTRACT

Mutations in the master hematopoietic transcription factor GATA1 are often associated with functional defects in erythropoiesis and megakaryopoiesis. In this study, we identified a novel GATA1 germline mutation (c.1162delGG, p.Leu387Leufs*62) in a patient with congenital anemia and occasional thrombocytopenia. The C-terminal GATA1, a rarely studied mutational region, undergoes frameshifting translation as a consequence of this double-base deletion mutation. To investigate the specific function and pathogenic mechanism of this mutant, in vitro mutant models of stable re-expression cells were generated. The mutation was subsequently validated to cause diminished transcriptional activity of GATA1 and defective differentiation of erythroid and megakaryocytes. Using proximity labeling and mass spectrometry, we identified selective alterations in the proximal protein networks of the mutant, revealing decreased binding to a set of normal GATA1-interaction proteins, including the essential co-factor FOG1. Notably, our findings further demonstrated enhanced recruitment of the protein arginine methyltransferase PRMT6, which mediates histone modification at H3R2me2a and represses transcription activity. We also found an enhanced binding of this mutant GATA1/PRMT6 complex to the transcriptional regulatory elements of GATA1's target genes. Moreover, treatment of the PRMT6 inhibitor MS023 could partially rescue the inhibited transcriptional and impaired erythroid differentiation caused by the GATA1 mutation. Taken together, our results provide molecular insights into erythropoiesis in which mutation leads to partial loss of GATA1 function, and the role of PRMT6 and its inhibitor MS023 in congenital anemia, highlighting PRMT6 binding as a negative factor of GATA1 transcriptional activity in aberrant hematopoiesis.


Subject(s)
GATA1 Transcription Factor , Germ-Line Mutation , Protein Binding , Protein-Arginine N-Methyltransferases , Humans , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Cell Differentiation/genetics , Erythropoiesis/genetics , Male , Female , Anemia/genetics
15.
Blood ; 143(13): 1282-1292, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38232308

ABSTRACT

ABSTRACT: As a functional component of erythrocyte hemoglobin, iron is essential for oxygen delivery to all tissues in the body. The liver-derived peptide hepcidin is the master regulator of iron homeostasis. During anemia, the erythroid hormone erythroferrone regulates hepcidin synthesis to ensure the adequate supply of iron to the bone marrow for red blood cell production. However, mounting evidence suggested that another factor may exert a similar function. We identified the hepatokine fibrinogen-like 1 (FGL1) as a previously undescribed suppressor of hepcidin that is induced in the liver in response to hypoxia during the recovery from anemia, and in thalassemic mice. We demonstrated that FGL1 is a potent suppressor of hepcidin in vitro and in vivo. Deletion of Fgl1 in mice results in higher hepcidin levels at baseline and after bleeding. FGL1 exerts its activity by directly binding to bone morphogenetic protein 6 (BMP6), thereby inhibiting the canonical BMP-SMAD signaling cascade that controls hepcidin transcription.


Subject(s)
Anemia , Hepcidins , Mice , Animals , Hepcidins/genetics , Hepcidins/metabolism , Anemia/genetics , Anemia/metabolism , Iron/metabolism , Liver/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Homeostasis
16.
Br J Nutr ; 131(2): 193-201, 2024 01 28.
Article in English | MEDLINE | ID: mdl-37605822

ABSTRACT

Fe-deficiency anaemia is a major public health concern in children under 5 years of age. TMPRSS6 gene, encoding matriptase-2 protein, is implicated in Fe homoeostasis and has been associated with anaemia and Fe status in various populations. The aim of this cross-sectional study was to investigate the associations between the single nucleotide polymorphism (SNP) TMPRSS6 rs855791 and biomarkers of anaemia and Fe deficiency in Brazilian children attending day care centres. A total of 163 children aged 6-42 months were evaluated. Socio-economic, demographic, biochemical, haematological, immunological and genotype data were collected. Multiple logistic and linear regressions with hierarchical selection were used to assess the effects of independent variables on categorised outcomes and blood marker concentrations. Minor allele (T) frequency of rs855791 was 0·399. Each copy of the T allele was associated with a 4·49-fold increased risk of developing anaemia (P = 0·005) and a 4·23-fold increased risk of Fe deficiency assessed by serum soluble transferrin receptor (sTfR) (P < 0·001). The dose of the T allele was associated with an increase of 0·18 mg/l in sTfR concentrations and reductions of 1·41 fl and 0·52 pg in mean corpuscular volume (MCV) and mean corpuscular haemoglobin (MCH), respectively. In conclusion, the T allele of SNP TMPRSS6 rs855791 was significantly associated with anaemia and Fe deficiency assessed by sTfR in Brazilian children attending day care centres. The effect was dose dependent, with each copy of the T allele being associated with lower MCV and MCH and higher concentrations of sTfR.


Subject(s)
Anemia, Iron-Deficiency , Anemia , Iron Deficiencies , Child, Preschool , Humans , Anemia/epidemiology , Anemia/genetics , Anemia, Iron-Deficiency/epidemiology , Anemia, Iron-Deficiency/genetics , Brazil/epidemiology , Cross-Sectional Studies , Day Care, Medical , Membrane Proteins/genetics , Polymorphism, Single Nucleotide , Receptors, Transferrin , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
18.
Am J Hematol ; 99(1): 12-20, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37867341

ABSTRACT

Ferritin is a hetero-oligomeric nanocage, composed of 24 subunits of two types, FTH1 and FTL. It protects the cell from excess reactive iron, by storing iron in its cavity. FTH1 is essential for the recruitment of iron into the ferritin nanocage and for cellular ferritin trafficking, whereas FTL contributes to nanocage stability and iron nucleation inside the cavity. Here we describe a female patient with a medical history of severe hypoferritinemia without anemia. Following inadequate heavy IV iron supplementation, the patient developed severe iron overload and musculoskeletal manifestations. However, her serum ferritin levels rose only to normal range. Genetic analyses revealed an undescribed homozygous variant of FTL (c.92A > G), which resulted in a Tyr31Cys substitution (FTLY31C ). Analysis of the FTL structure predicted that the Y31C mutation will reduce the variant's stability. Expression of the FTLY31C variant resulted in significantly lower cellular ferritin levels compared with the expression of wild-type FTL (FTLWT ). Proteasomal inhibition significantly increased the initial levels of FTLY31C , but could not protect FTLY31C subunits from successive degradation. Further, variant subunits successfully incorporated into hetero-polymeric nanocages in the presence of sufficient levels of FTH1. However, FTLY31C subunits poorly assembled into nanocages when FTH1 subunit levels were low. These results indicate an increased susceptibility of unassembled monomeric FTLY31C subunits to proteasomal degradation. The decreased cellular assembly of FTLY31C -rich nanocages may explain the low serum ferritin levels in this patient and emphasize the importance of a broader diagnostic approach of hypoferritinemia without anemia, before IV iron supplementation.


Subject(s)
Anemia , Apoferritins , Iron Deficiencies , Iron Overload , Female , Humans , Anemia/genetics , Apoferritins/genetics , Apoferritins/metabolism , Ferritins , Iron/metabolism , Iron Deficiencies/genetics , Iron Overload/genetics
19.
ESC Heart Fail ; 11(1): 299-305, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37984882

ABSTRACT

AIMS: Observational studies have suggested that anaemia is associated with an increased risk of heart failure (HF). But the potential causal association is not clear. We aimed to investigate the association between anaemia and HF risk. METHODS AND RESULTS: A Mendelian randomization (MR) analysis was performed to confirm the causal association of anaemia with the risk of HF and left ventricular structure and function. Furthermore, a reverse-direction MR analyses was conducted to assess the causal effect of HF on anaemia. The MR analysis indicated that genetically predicted anaemia is associated with the increased risk of HF (meta: odd ratio (OR) = 1.12; 95% confidence interval (CI) [1.04, 1.20]; P = 0.002), and left ventricular mass index (ß = 1.051; 95% CI [0.384, 1.718]; P = 0.002), left ventricular mass (ß = 2.063; 95% CI [0.578, 3.547]; P = 0.006), left atrial minimum volume (ß = 0.076; 95% CI [0.008, 0.143]; P = 0.028), and left atrial maximum volume (ß = 0.090; 95% CI [0.023, 0.157]; P = 0.009). In the reverse-direction MR analyses, we found that genetic susceptibility to HF was significantly associated with the increased risk of anaemia (meta: OR = 1.40; 95% CI [1.24, 1.59]; P = 1.79 × 10-7 ). CONCLUSIONS: This MR study supports the genetic evidence that there is bidirectional causality between anaemia and the risk of HF as well as anaemia may cause left ventricular hypertrophy and enlargement of the left atrium. Considering the adverse causal effects between the two diseases, more attention should be paid to the prevention and treatment of anaemia in patients with HF.


Subject(s)
Anemia , Heart Failure , Humans , Ventricular Function, Left , Mendelian Randomization Analysis , Heart Failure/complications , Heart Failure/genetics , Hypertrophy, Left Ventricular/complications , Hypertrophy, Left Ventricular/genetics , Anemia/complications , Anemia/epidemiology , Anemia/genetics
20.
Chin J Traumatol ; 27(1): 27-33, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37423837

ABSTRACT

PURPOSE: Dabigatran is usually prescribed in recommended doses without monitoring of the blood coagulation for the prevention of venous thromboembolism after joint arthroplasty. ABCB1 is a key gene in the metabolism of dabigatran etexilate. Its allele variants are likely to play a pivotal role in the occurrence of hemorrhagic complications. METHODS: The prospective study included 127 patients with primary knee osteoarthritis undergoing total knee arthroplasty. Patients with anemia and coagulation disorders, elevated transaminase and creatinine levels as well as already receiving anticoagulant and antiplatelet therapy were excluded from the study. The association of ABCB1 gene polymorphisms rs1128503, rs2032582, rs4148738 with anemia as the outcome of dabigatran therapy was evaluated by single-nucleotide polymorphism analysis with a real-time polymerase chain reaction assay and laboratory blood tests. The beta regression model was used to predict the effect of polymorphisms on the studied laboratory markers. The probability of the type 1 error (p) was less than 0.05 was considered statistically significant. BenjaminiHochberg was used to correct for significance levels in multiple hypothesis tests. All calculations were performed using Rprogramming language v3.6.3. RESULTS: For all polymorphisms there was no association with the level of platelets, protein, creatinine, alanine transaminase, prothrombin, international normalized ratio, activated partial thromboplastin time and fibrinogen. Carriers of rs1128503 (TT) had a significant decrease of hematocrit (p = 0.001), red blood count and hemoglobin (p = 0.015) while receiving dabigatran therapy during the postoperative period compared to the CC, CT. Carriers of rs2032582 (TT) had a significant decrease of hematocrit (p = 0.001), red blood count and hemoglobin (p = 0.006) while receiving dabigatran therapy during the postoperative period compared to the GG, GT phenotypes. These differences were not observed in carriers of rs4148738. CONCLUSION: It might be necessary to reconsider thromboprophylaxis with dabigatran in carriers of rs1128503 (TT) or rs2032582 (TT) polymorphisms in favor of other new oral anticoagulants. The long-term implication of these findings would be the reduction of bleeding complications after total joint arthroplasty.


Subject(s)
Anemia , Anticoagulants , Arthroplasty, Replacement, Knee , Dabigatran , Venous Thromboembolism , Humans , Anemia/genetics , Anemia/prevention & control , Anticoagulants/therapeutic use , Arthroplasty, Replacement, Knee/adverse effects , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Creatinine , Dabigatran/therapeutic use , Hemoglobins , Polymorphism, Genetic , Prospective Studies , Venous Thromboembolism/genetics , Venous Thromboembolism/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL