Matriptase-2 is essential for hepcidin repression during fetal life and postnatal development in mice to maintain iron homeostasis

Blood ◽  
2014 ◽  
Vol 124 (3) ◽  
pp. 441-444 ◽  
Author(s):  
Alexandra Willemetz ◽  
Anne Lenoir ◽  
Jean-Christophe Deschemin ◽  
Carlos Lopez-Otin ◽  
Andrew J. Ramsay ◽  
...  

Key Points Matriptase-2 is required for hepcidin suppression in fetuses and neonates to ensure iron mobilization to avoid iron deficiency and anemia. In matriptase-2–deficient mice, the constant developmental hyperhepcidinemia is not explained by increased level of hepatic hemojuvelin.

Blood ◽  
2011 ◽  
Vol 118 (3) ◽  
pp. 736-746 ◽  
Author(s):  
Alessia Pagani ◽  
Antonella Nai ◽  
Gianfranca Corna ◽  
Lidia Bosurgi ◽  
Patrizia Rovere-Querini ◽  
...  

AbstractHepcidin is an antimicrobial peptide that controls systemic iron homeostasis. Hepcidin binding to its receptor ferroportin reduces iron availability, thus controlling microbial growth. In parallel it triggers an anti-inflammatory response in macrophages. Hepcidin is transcriptionally regulated by iron, through the bone morphogenetic protein–son of mothers against decapentaplegic (BMP-SMAD) pathway and by inflammation, through IL6-mediated STAT3 signaling. To investigate the mechanisms linking iron and inflammation, we treated C57BL/6 iron-deficient mice with a sublethal dose of lipopolysaccharide (LPS) and analyzed their inflammatory response in comparison with controls. We show that iron-deprived mice have a proinflammatory condition, exacerbated by LPS treatment leading to increased IL6 and TNFα mRNA in liver and spleen macrophages, and increased serum IL6 (482.29 ± 205.59 pg/mL) versus controls (69.01 ± 17.52 pg/mL; P < .05). Hepcidin was undetectable in iron-deficient mice but pretreatment with hepcidin normalized their response to LPS. Tmprss6−/− mice, characterized by iron deficiency and high hepcidin, show a blunted inflammatory response when challenged with LPS. Our data support a model in which the lack of hepcidin is responsible of the high inflammatory response to LPS in iron deficiency. The proinflammatory status associated with chronic iron deficiency could explain the resistance to infection seen in this condition.


2021 ◽  
Author(s):  
Wei‐Jian Sun ◽  
Jiu‐Cheng Zhang ◽  
Xing‐Long Ji ◽  
Zi‐Quan Feng ◽  
Xun Wang ◽  
...  

Blood ◽  
2017 ◽  
Vol 130 (20) ◽  
pp. 2224-2228 ◽  
Author(s):  
Elizabeth J. Haining ◽  
Deya Cherpokova ◽  
Karen Wolf ◽  
Isabelle C. Becker ◽  
Sarah Beck ◽  
...  

Key Points An inactivating point mutation in the hemITAM motif of murine CLEC-2 reproduces the lymphatic defects seen in CLEC-2–deficient mice. CLEC-2 contributes to thrombus stability in vivo independently of hemITAM signaling.


Blood ◽  
2014 ◽  
Vol 123 (5) ◽  
pp. 615-624 ◽  
Author(s):  
Nicholas J. Kassebaum ◽  
Rashmi Jasrasaria ◽  
Mohsen Naghavi ◽  
Sarah K. Wulf ◽  
Nicole Johns ◽  
...  

Key Points Anemia accounted for 8.8% of the total disability from all conditions in 2010. Children <5 years and women still have the highest burden. Although iron-deficiency anemia is the most common etiology globally, other leading causes of anemia vary widely by geography, age, and sex.


2011 ◽  
Vol 47 (3) ◽  
pp. 151-160 ◽  
Author(s):  
Jennifer L. McCown ◽  
Andrew J. Specht

Iron is an essential element for nearly all living organisms and disruption of iron homeostasis can lead to a number of clinical manifestations. Iron is used in the formation of both hemoglobin and myoglobin, as well as numerous enzyme systems of the body. Disorders of iron in the body include iron deficiency anemia, anemia of inflammatory disease, and iron overload. This article reviews normal iron metabolism, disease syndromes of iron imbalance, diagnostic testing, and treatment of either iron deficiency or excess. Recent advances in diagnosing iron deficiency using reticulocyte indices are reviewed.


2018 ◽  
Vol 115 (12) ◽  
pp. 3000-3005 ◽  
Author(s):  
Benjamin H. Hudson ◽  
Andrew T. Hale ◽  
Ryan P. Irving ◽  
Shenglan Li ◽  
John D. York

Sulfur assimilation is an evolutionarily conserved pathway that plays an essential role in cellular and metabolic processes, including sulfation, amino acid biosynthesis, and organismal development. We report that loss of a key enzymatic component of the pathway, bisphosphate 3′-nucleotidase (Bpnt1), in mice, both whole animal and intestine-specific, leads to iron-deficiency anemia. Analysis of mutant enterocytes demonstrates that modulation of their substrate 3′-phosphoadenosine 5′-phosphate (PAP) influences levels of key iron homeostasis factors involved in dietary iron reduction, import and transport, that in part mimic those reported for the loss of hypoxic-induced transcription factor, HIF-2α. Our studies define a genetic basis for iron-deficiency anemia, a molecular approach for rescuing loss of nucleotidase function, and an unanticipated link between nucleotide hydrolysis in the sulfur assimilation pathway and iron homeostasis.


Blood ◽  
2014 ◽  
Vol 123 (23) ◽  
pp. 3646-3650 ◽  
Author(s):  
Sara Zumerle ◽  
Jacques R. R. Mathieu ◽  
Stéphanie Delga ◽  
Mylène Heinis ◽  
Lydie Viatte ◽  
...  

Key Points Liver-specific hepcidin KO mice fully recapitulate the severe iron overload phenotype observed in the total KO mice. The hepcidin produced by hepatocytes is the main regulator of body iron homeostasis.


Blood ◽  
2017 ◽  
Vol 129 (4) ◽  
pp. 405-414 ◽  
Author(s):  
Susanna Canali ◽  
Kimberly B. Zumbrennen-Bullough ◽  
Amanda B. Core ◽  
Chia-Yu Wang ◽  
Manfred Nairz ◽  
...  

Key Points Endothelial Bmp6 conditional knockout mice exhibit hemochromatosis, whereas hepatocyte and macrophage Bmp6 conditional knockout mice do not. Our data support a model in which EC Bmp6 has paracrine actions on hepatocyte hemojuvelin to regulate hepcidin production.


Endocrinology ◽  
2021 ◽  
Author(s):  
Hannah Roberts ◽  
Andrew G Woodman ◽  
Kelly J Baines ◽  
Mariyan J Jeyarajah ◽  
Stephane L Bourque ◽  
...  

Abstract Iron deficiency occurs when iron demands chronically exceed intake, and is prevalent in pregnant women. Iron deficiency during pregnancy poses major risks for the baby, including fetal growth restriction and long-term health complications. The placenta serves as the interface between a pregnant mother and her baby, and ensures adequate nutrient provisions for the fetus. Thus, maternal iron deficiency may impact fetal growth and development by altering placental function. We used a rat model of diet-induced iron deficiency to investigate changes in placental growth and development. Pregnant Sprague-Dawley rats were fed either a low-iron or iron-replete diet starting two weeks before mating. Compared to controls, both maternal and fetal hemoglobin were reduced in dams fed low-iron diets. Iron deficiency decreased fetal liver and body weight, but not brain, heart or kidney weight. Placental weight was increased in iron deficiency, due primarily to expansion of the placental junctional zone. The stimulatory effect of iron deficiency on junctional zone development was recapitulated in vitro, as exposure of rat trophoblast stem cells to the iron chelator deferoxamine increased differentiation toward junctional zone trophoblast subtypes. Gene expression analysis revealed 464 transcripts changed at least 1.5-fold (P&lt;0.05) in placentas from iron-deficient dams, including altered expression of genes associated with oxygen transport and lipoprotein metabolism. Expression of genes associated with iron homeostasis was unchanged despite differences in levels of their encoded proteins. Our findings reveal robust changes in placentation during maternal iron deficiency, which could contribute to the increased risk of fetal distress in these pregnancies.


2020 ◽  
Author(s):  
Xin Zhang ◽  
Xiaojin Zhou ◽  
Suzhen Li ◽  
Jiaxing Huang ◽  
Sen Pang ◽  
...  

Abstract Background: Nicotianamine (NA) serves as not only the major chelator for iron transport but also the intermediate for synthesizing mugineic acid family phytosiderophores (MAs) which are secreted by graminaceous plants for Fe uptake. Therefore, the production and secretion of MAs are key steps for maintaining iron homeostasis in plants. Nicotianamine aminotransferase (NAAT), 2’-deoxymugineic acid synthase (DMAS), MAs efflux transporter (TOM), and efflux transporter of NA (ENA) were identified to be involved in these processes in rice and barley, whereas little systematic study has been performed in maize (Zea mays.L). Results: Here, we identified five ZmNAAT, nine ZmDMAS, eleven ZmTOM, and two ZmENA genes in maize by genome mining. RNA-sequencing (RNA-seq) and quantitative real-time PCR (qRT-PCR) analysis revealed that the expression of these genes exhibited diverse tissue specificity and different responses to environmental iron conditions. Moreover, the expression patterns were related to their evolution relationships. In particular, the ZmNAAT family can be classified into two subgroups, with one group showed inhibited expression in root under iron excess status and another subclass were repressed in shoot under both iron deficiency and excess. Likewise, the expression of ZmDMAS1 was stimulated under iron deficiency, while the remaining genes fell into two sub-clades with different expression patterns. Significant up-regulation of ZmTOM1, ZmTOM3 and ZmENA1 were observed under iron starvation, while ZmTOM2 was induced under both iron-excess and deficiency. These results reflect changing demands for the synthesis and secretion of NA/MAs to balance iron homeostasis under fluctuating conditions. All the examined ZmNAAT and ZmDMAS proteins localized in cytoplasm, while plasma and tonoplast membrane, endomembrane, and vesicle localization were observed for ZmTOM and ZmENA proteins. These results indicate that ZmTOM and ZmENA proteins may contribute to not only intercellular export but also intracellular sequestration of NA and MAs to facilitate iron homeostasis. Conclusions: Our results suggest that different gene expression profiles and subcellular localization of ZmNAAT, ZmDMAS, ZmTOM, and ZmENA members may enable dedicate regulation of NA and phytosiderophores (PS) metabolism, shedding light on the understanding of iron-homeostasis in maize. Additionally, we also provided candidate genes for breeding iron-rich maize varieties.


Sign in / Sign up

Export Citation Format

Share Document