Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 252
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Blood ; 143(23): 2433-2437, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38518102

ABSTRACT

ABSTRACT: Iron-mediated induction of bone morphogenetic protein (BMP)6 expression by liver endothelial cells is essential for iron homeostasis regulation. We used multiple dietary and genetic mouse cohorts to demonstrate a minor functional role for the metal-ion transporter ZIP8 in regulating BMP6 expression under high-iron conditions.


Subject(s)
Bone Morphogenetic Protein 6 , Cation Transport Proteins , Iron , Animals , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/genetics , Mice , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Iron/metabolism , Endothelial Cells/metabolism , Mice, Knockout , Gene Expression Regulation , Liver/metabolism , Mice, Inbred C57BL , Homeostasis
3.
Blood ; 142(15): 1312-1322, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37478395

ABSTRACT

Hepcidin is the master regulator of systemic iron homeostasis. The bone morphogenetic protein (BMP) signaling pathway is a critical regulator of hepcidin expression in response to iron and erythropoietic drive. Although endothelial-derived BMP6 and BMP2 ligands have key functional roles as endogenous hepcidin regulators, both iron and erythropoietic drives still regulate hepcidin in mice lacking either or both ligands. Here, we used mice with an inactivating Bmp5 mutation (Bmp5se), either alone or together with a global or endothelial Bmp6 knockout, to investigate the functional role of BMP5 in hepcidin and systemic iron homeostasis regulation. We showed that Bmp5se-mutant mice exhibit hepcidin deficiency at age 10 days, blunted hepcidin induction in response to oral iron gavage, and mild liver iron loading when fed on a low- or high-iron diet. Loss of 1 or 2 functional Bmp5 alleles also leads to increased iron loading in Bmp6-heterozygous mice and more profound hemochromatosis in global or endothelial Bmp6-knockout mice. Moreover, double Bmp5- and Bmp6-mutant mice fail to induce hepcidin in response to long-term dietary iron loading. Finally, erythroferrone binds directly to BMP5 and inhibits BMP5 induction of hepcidin in vitro. Although erythropoietin suppresses hepcidin in Bmp5se-mutant mice, it fails to suppress hepcidin in double Bmp5- and Bmp6-mutant males. Together, these data demonstrate that BMP5 plays a functional role in hepcidin and iron homeostasis regulation, particularly under conditions in which BMP6 is limited.


Subject(s)
Hemochromatosis , Hepcidins , Animals , Male , Mice , Bone Morphogenetic Protein 6/metabolism , Hemochromatosis/genetics , Hepcidins/genetics , Hepcidins/metabolism , Homeostasis , Iron/metabolism , Liver/metabolism , Mice, Knockout
4.
Blood ; 141(3): 271-284, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36351237

ABSTRACT

Homeostatic adaptation to systemic iron overload involves transcriptional induction of bone morphogenetic protein 6 (BMP6) in liver sinusoidal endothelial cells (LSECs). BMP6 is then secreted to activate signaling of the iron hormone hepcidin (HAMP) in neighboring hepatocytes. To explore the mechanism of iron sensing by LSECs, we generated TfrcTek-Cre mice with endothelial cell-specific ablation of transferrin receptor 1 (Tfr1). We also used control Tfrcfl/fl mice to characterize the LSEC-specific molecular responses to iron using single-cell transcriptomics. TfrcTek-Cre animals tended to have modestly increased liver iron content (LIC) compared with Tfrcfl/fl controls but expressed physiological Bmp6 and Hamp messenger RNA (mRNA). Despite a transient inability to upregulate Bmp6, they eventually respond to iron challenges with Bmp6 and Hamp induction, yet occasionally to levels slightly lower relative to LIC. High dietary iron intake triggered the accumulation of serum nontransferrin bound iron (NTBI), which significantly correlated with liver Bmp6 and Hamp mRNA levels and elicited more profound alterations in the LSEC transcriptome than holo-transferrin injection. This culminated in the robust induction of Bmp6 and other nuclear factor erythroid 2-related factor 2 (Nrf2) target genes, as well as Myc target genes involved in ribosomal biogenesis and protein synthesis. LSECs and midzonal hepatocytes were the most responsive liver cells to iron challenges and exhibited the highest expression of Bmp6 and Hamp mRNAs, respectively. Our data suggest that during systemic iron overload, LSECs internalize NTBI, which promotes oxidative stress and thereby transcriptionally induces Bmp6 via Nrf2. Tfr1 appears to contribute to iron sensing by LSECs, mostly under low iron conditions.


Subject(s)
Iron Overload , Iron , Mice , Animals , Iron/metabolism , Transferrin/metabolism , Endothelial Cells/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , NF-E2-Related Factor 2 , Hepatocytes/metabolism , Liver/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Iron Overload/genetics , Iron Overload/metabolism , RNA, Messenger/metabolism
5.
Cell Tissue Res ; 396(1): 103-117, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403744

ABSTRACT

The formation of the epiretinal fibrotic membrane by retinal pigment epithelial (RPE) cells is a primary pathological change for proliferative vitreoretinopathy (PVR). Bone morphogenetic protein 6 (BMP6) is an antifibrogenic factor in various cells. To date, it is still unknown whether BMP6 can interfere with the fibrogenesis of RPE cells during the progression of PVR. This work aimed to address the relationship between BMP6 and transforming growth factor-ß2 (TGF-ß2)-elicited fibrogenesis of RPE cells, an experimental model for studying PVR in vitro. The BMP6 level was down-regulated, while the TGF-ß2 level was up-regulated in the vitreous humor of PVR patients. The BMP6 level was down-regulated in human RPE cells challenged with TGF-ß2. The treatment of RPE cells with TGF-ß2 resulted in significant increases in proliferation, migration, epithelial-to-mesenchymal transition (EMT), and extracellular matrix (ECM) remodelling. These effects were found to be inhibited by the overexpression of BMP6 or exacerbated by the knockdown of BMP6. BMP6 overexpression reduced the phosphorylation of p38 and JNK in TGF-ß2-stimulated RPE cells, while BMP6 knockdown showed the opposite effects. The inhibition of p38 or JNK partially reversed the BMP6-silencing-induced promoting effects on TGF-ß2-elicited fibrogenesis in RPE cells. Taken together, BMP6 demonstrates the ability to counteract the proliferation, migration, EMT, and ECM remodelling of RPE cells induced by TGF-ß2. This is achieved through the regulation of the p38 and JNK MAPK pathways. These findings imply a potential connection between BMP6 and PVR, and highlight the potential application of BMP6 in therapeutic interventions for PVR.


Subject(s)
Vitreoretinopathy, Proliferative , Humans , Vitreoretinopathy, Proliferative/drug therapy , Vitreoretinopathy, Proliferative/metabolism , Vitreoretinopathy, Proliferative/pathology , Retinal Pigment Epithelium , Transforming Growth Factor beta2/pharmacology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/therapeutic use , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/therapeutic use , Epithelial-Mesenchymal Transition , Epithelial Cells/metabolism , Retinal Pigments/metabolism , Retinal Pigments/pharmacology , Retinal Pigments/therapeutic use , Cell Movement
6.
J Assist Reprod Genet ; 41(1): 31-48, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37930517

ABSTRACT

PURPOSE: To evaluate whether PTX3 is differentially expressed in the granulosa lutein cells derived from women with PCOS and whether BMP6 can regulate the expression of PTX3 in hGL cells. METHODS: The expression levels of BMP6 and PTX3 in granulosa lutein cells were evaluated by RT-qPCR. The correlation between the expression levels of BMP6 /PTX3 and oocyte quality indexes were analyzed using clinical samples. The cells were incubated with BMP6 at different concentrations and times to check the expression of PTX3 in KGN cells. TGF-ß type I inhibitors and small interfering RNA targeting ALK2/3/6,SMAD1/5/8 and SMAD4 were used to study the involvement of SMAD dependent pathways in KGN cells. RESULTS: The levels of BMP6 in hGL cells were negatively correlated with the corresponding oocyte maturation rate and high-quality embryo rate, whereas the levels of PTX3 were positively correlated with the corresponding oocyte maturation rate in PCOS. Additionally, the in vitro cell cultured results showed BMP6 significantly inhibited the expression of PTX3 in KGN cells. Furthermore, using a dual inhibition approach (kinase inhibitors and small interfering RNAs), we identified the ALK2/ALK3 type I receptors and BMPR2/ACVR2A type II receptors and the downstream SMAD1/SMAD5-SMAD4 signaling pathway were responsible for the BMP6-induced cellular activities in KGN cells. CONCLUSIONS: The suppressive effect of BMP6 on PTX3 was mediated by ALK2/ALK3 type I receptors and BMPR2/ACVR2A type II receptors in granulosa cells through the SMAD1/5-SMAD4 dependent signaling pathway in PCOS.Our findings provides new insights into the understanding of the pathogenesis of PCOS-related ovulatory disorders.


Subject(s)
C-Reactive Protein , Luteal Cells , Polycystic Ovary Syndrome , Serum Amyloid P-Component , Female , Humans , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein Receptors, Type II/genetics , Down-Regulation/genetics , Granulosa Cells/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism
7.
Stem Cells ; 40(9): 843-856, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35759955

ABSTRACT

Quiescent hair follicle stem cells (HFSCs) reside in specialized bulge niche where they undergo activation and differentiation upon sensing niche-dependent signals during hair follicle (HF) homeostasis and wound repair. The underlying mechanism of HFSCs and bulge niche maintenance is poorly understood. Our previous study has reported that a transcription factor, forkhead box P1 (Foxp1), functions to maintain the quiescence of HFSCs. Here, we further discovered that forkhead box P4 (Foxp4), a close family member of Foxp1, had similar expression profiles in various components of HFs and formed a complex with Foxp1 in vitro and in vivo. The HF-specific deficiency of Foxp4 resulted in the precocious activation of HFSCs during hair cycles. In contrast to single Foxp1 or Foxp4 conditional knockout (cKO) mice, Foxp1/4 double cKO exerted an additive effect in the spectrum and severity of phenotypes in HFSC activation, hair cycling acceleration and hair loss, coupled with remarkable downregulation of fibroblast growth factor 18 (Fgf18) and bone morphogenetic protein 6 (Bmp6) expression in bulge cells. In addition, the double KO of Foxp1/4 induced the apoptosis of K6-positive (K6+) inner bulge cells, a well-established stem cell (SC) niche, thus resulting in the destruction of the bulge SC niche and recurrent hair loss. Our investigation reveals the synergistic role of Foxp1/4 in sustaining K6+ niche cells for the quiescence of HFSCs.


Subject(s)
Bone Morphogenetic Protein 6 , Stem Cell Niche , Alopecia/metabolism , Animals , Apoptosis/genetics , Bone Morphogenetic Protein 6/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Hair Follicle , Mice , Repressor Proteins/metabolism
8.
J Biol Chem ; 297(6): 101378, 2021 12.
Article in English | MEDLINE | ID: mdl-34740612

ABSTRACT

Liver sinusoidal endothelial cell-derived bone morphogenetic protein 6 (BMP6) and the BMP6-small mothers against decapentaplegic homolog (SMAD) signaling pathway are essential for the expression of hepcidin, the secretion of which is considered the systemic master switch of iron homeostasis. However, there are continued controversies related to the strong and direct suppressive effect of iron on hepatocellular hepcidin in vitro in contrast to in vivo conditions. Here, we directly studied the crosstalk between endothelial cells (ECs) and hepatocytes using in vitro coculture models that mimic hepcidin signaling in vivo. Huh7 cells were directly cocultured with ECs, and EC conditioned media (CM) were also used to culture Huh7 cells and primary mouse hepatocytes. To explore the reactions of ECs to surrounding iron, they were grown in the presence of ferric ammonium citrate and heme, two iron-containing molecules. We found that both direct coculture with ECs and EC-CM significantly increased hepcidin expression in Huh7 cells. The upstream SMAD pathway, including phosphorylated SMAD1/5/8, SMAD1, and inhibitor of DNA binding 1, was induced by EC-CM, promoting hepcidin expression. Efficient blockage of this EC-mediated hepcidin upregulation by an inhibitor of the BMP6 receptor ALK receptor tyrosine kinase 2/3 or BMP6 siRNA identified BMP6 as a major hepcidin regulator in this coculture system, which highly fits the model of hepcidin regulation by iron in vivo. In addition, EC-derived BMP6 and hepcidin were highly sensitive to levels of not only ferric iron but also heme as low as 500 nM. We here establish a hepatocyte-endothelial coculture system to fully recapitulate iron regulation by hepcidin using EC-derived BMP6.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Endothelial Cells/metabolism , Hepatocytes/metabolism , Iron/metabolism , Animals , Cell Line , Coculture Techniques , Gene Silencing , Hepcidins/genetics , Hepcidins/metabolism , Humans , Male , Mice , Signal Transduction
9.
Hepatology ; 74(4): 2186-2200, 2021 10.
Article in English | MEDLINE | ID: mdl-33982327

ABSTRACT

BACKGROUND AND AIMS: TGFß/bone morphogenetic protein (BMP) signaling in the liver plays a critical role in liver disease. Growth factors, such as BMP2, BMP6, and TGFß1, are released from LSECs and signal in a paracrine manner to hepatocytes and hepatic stellate cells to control systemic iron homeostasis and fibrotic processes, respectively. The misregulation of the TGFß/BMP pathway affects expression of the iron-regulated hormone hepcidin, causing frequent iron overload and deficiency diseases. However, whether LSEC-secreted factors can act in an autocrine manner to maintain liver homeostasis has not been addressed so far. APPROACH AND RESULTS: We analyzed publicly available RNA-sequencing data of mouse LSECs for ligand-receptor interactions and identified members of the TGFß family (BMP2, BMP6, and TGFß1) as ligands with the highest expression levels in LSECs that may signal in an autocrine manner. We next tested the soluble factors identified through in silico analysis in optimized murine LSEC primary cultures and mice. Exposure of murine LSEC primary cultures to these ligands shows that autocrine responses to BMP2 and BMP6 are blocked despite high expression levels of the required receptor complexes partially involving the inhibitor FK-506-binding protein 12. By contrast, LSECs respond efficiently to TGFß1 treatment, which causes reduced expression of BMP2 through activation of activin receptor-like kinase 5. CONCLUSIONS: These findings reveal that TGFß1 signaling is functionally interlinked with BMP signaling in LSECs, suggesting druggable targets for the treatment of iron overload diseases associated with deficiency of the BMP2-regulated hormone hepcidin, such as hereditary hemochromatosis, ß-thalassemia, and chronic liver diseases.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 6/metabolism , Hepcidins/metabolism , Iron/metabolism , Liver Cirrhosis , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Drug Discovery , Endothelial Cells/metabolism , Gene Expression Regulation , Hepatic Stellate Cells , Hepatocytes/metabolism , Homeostasis , Iron Overload/drug therapy , Iron Overload/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice
10.
Am J Hematol ; 97(12): 1548-1559, 2022 12.
Article in English | MEDLINE | ID: mdl-36069607

ABSTRACT

Systemic iron homeostasis is regulated by the hepatic hormone hepcidin to balance meeting iron requirements while limiting toxicity from iron excess. Iron-mediated induction of bone morphogenetic protein (BMP) 6 is a central mechanism for regulating hepcidin production. Liver endothelial cells (LECs) are the main source of endogenous BMP6, but how they sense iron to modulate BMP6 transcription and thereby hepcidin is uncertain. Here, we investigate the role of endothelial cell transferrin receptor 1 (TFR1) in iron uptake, BMP6 regulation, and systemic iron homeostasis using primary LEC cultures and endothelial Tfrc (encoding TFR1) knockout mice. We show that intracellular iron regulates Bmp6 expression in a cell-autonomous manner, and TFR1 mediates iron uptake and Bmp6 expression by holo-transferrin in primary LEC cultures. In addition, endothelial Tfrc knockout mice exhibit altered iron homeostasis compared with littermate controls when fed a limited iron diet, as evidenced by increased liver iron and inappropriately low Bmp6 and hepcidin expression relative to liver iron. However, endothelial Tfrc knockout mice have a similar iron phenotype compared to littermate controls when fed an iron-rich standard diet. Finally, ferritin and non-transferrin bound iron (NTBI) are additional sources of iron that mediate Bmp6 induction in primary LEC cultures via TFR1-independent mechanisms. Together, our data demonstrate a minor functional role for endothelial cell TFR1 in iron uptake, BMP6 regulation, and hepatocyte hepcidin regulation under iron limiting conditions, and suggest that ferritin and/or NTBI uptake by other transporters have a dominant role when iron availability is high.


Subject(s)
Hepcidins , Iron , Mice , Animals , Hepcidins/genetics , Hepcidins/metabolism , Iron/metabolism , Endothelial Cells/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Homeostasis , Hepatocytes/metabolism , Ferritins , Transferrin/metabolism , Mice, Knockout
11.
Growth Factors ; 39(1-6): 1-13, 2021.
Article in English | MEDLINE | ID: mdl-34706618

ABSTRACT

The BMP6 protein (Bone Morphogenetic Protein 6) is part of the superfamily of transforming growth factor-beta (TGF-ß) ligands, participates in iron homeostasis, inhibits invasion by increasing adhesions and cell-cell type interactions and induces angiogenesis directly on vascular endothelial cells. BMP6 is coded by a tumor suppressor gene whose subexpression is related to the development and cancer progression; during neoplastic processes, methylation is the main mechanism by which gene silencing occurs. This work presents a review on the role of BMP6 protein in breast cancer (BC) and other types of cancer. The studies carried out to date suggest the participation of the BMP6 protein in the epithelial-mesenchymal transition (EMT) phenotype, cell growth and proliferation; however, these processes are affected in a variable way in the different types of cancer, the methylated CpG sites in BMP6 gene promoter, as well as the interaction with other proteins could be the cause of such variation.


Subject(s)
Bone Morphogenetic Protein 6 , Breast Neoplasms , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Transforming Growth Factor beta/metabolism
12.
Angiogenesis ; 24(1): 129-144, 2021 02.
Article in English | MEDLINE | ID: mdl-33021694

ABSTRACT

The BMP/TGFß-Smad, Notch and VEGF signaling guides formation of endothelial tip and stalk cells. However, the crosstalk of bone morphogenetic proteins (BMPs) and vascular endothelial growth factor receptor 2 (VEGFR2) signaling has remained largely unknown. We demonstrate that BMP family members regulate VEGFR2 and Notch signaling, and act via TAZ-Hippo signaling pathway. BMPs were found to be regulated after VEGF gene transfer in C57/Bl6 mice and in a porcine myocardial ischemia model. BMPs 2/4/6 were identified as endothelium-specific targets of VEGF. BMP2 modulated VEGF-mediated endothelial sprouting via Delta like Canonical Notch Ligand 4 (DLL4). BMP6 modulated VEGF signaling by regulating VEGFR2 expression and acted via Hippo signaling effector TAZ, known to regulate cell survival/proliferation, and to be dysregulated in cancer. In a matrigel plug assay in nude mice BMP6 was further demonstrated to induce angiogenesis. BMP6 is the first member of BMP family found to directly regulate both Hippo signaling and neovessel formation. It may thus serve as a target in pro/anti-angiogenic therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Protein 6/metabolism , Endothelial Cells/metabolism , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Base Sequence , Bone Morphogenetic Protein 2/metabolism , Cell Hypoxia , Cell Nucleus/metabolism , Hippo Signaling Pathway , Humans , Male , Mice, Inbred C57BL , Models, Biological , Protein Transport , Swine , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Neurochem Res ; 46(5): 1224-1238, 2021 May.
Article in English | MEDLINE | ID: mdl-33835366

ABSTRACT

The hormone hepcidin plays a central role in controlling iron homeostasis. Iron-mediated hepcidin synthesis is triggered via the BMP/SMAD pathway. At inflammation, mainly IL-6 pro-inflammatory cytokine mediates the regulation of hepcidin via the JAK/STAT signalling pathway. Microglial cells of the central nervous system are able to recognize a broad spectrum of pathogens via toll-like receptors and initiate inflammatory response. Although the regulation of hepcidin synthesis is well described in many tissues, little is known about the inflammation mediated hepcidin regulation in microglia. In this study, we investigated the pathways, which are involved in HAMP regulation in BV2 microglia due to inflammatory mediators and the possible relationships between the iron regulatory pathways. Our results showed that IL-6 produced by resting BV2 cells was crucial in maintaining the basal HAMP expression and hepcidin secretion. It was revealed that IL-6 neutralization decreased both STAT3 and SMAD1/5/9 phosphorylation suggesting that IL-6 proinflammatory cytokine is necessary to maintain SMAD1/5/9 activation. We revealed that IL-6 influences BMP6 and TMPRSS6 protein levels, moreover it modified TfR2 expression, as well. In this study, we revealed that BV2 microglia increased their hepcidin secretion upon IL-6 neutralization although the major regulatory pathways were inhibited. Based on our results it seems that both at inflammation and at normal condition the absence of IL-6 triggered HAMP transcription and hepcidin secretion via the NFκB pathway and possibly by the autocrine effect of TNFα cytokine on BV2 microglia.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Hepcidins/metabolism , Interleukin-6/metabolism , Membrane Proteins/metabolism , Microglia/metabolism , Receptors, Transferrin/metabolism , Serine Endopeptidases/metabolism , Animals , Cell Line , Cell Survival/drug effects , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Microglia/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/metabolism , Teichoic Acids/pharmacology
14.
Cell Mol Biol (Noisy-le-grand) ; 67(3): 69-75, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34933731

ABSTRACT

Gender determination, in addition to having special value to parents, has particular importance in sex-linked diseases. This study aimed to investigate the cellular indicators (i.e. BMP-6 protein and PPAR? protein expression levels in granulosa cells) and the physiological indicators on gender determination. For this purpose, on 68 infertile patients referred to the clinic, ovarian stimulation was performed by different protocols and then ruptured by different HCG. Follow-up of patients was performed after they became pregnant after five months. U/S was done for knowing the gender of the baby then after labor rechecked another time. Also, granulosa-luteal cells (GLCs) were isolated from the follicular fluid of 68 women participating in the study. BMP-6 protein and PPAR? protein were measured using Western blotting. Results showed that the total number of delivered babies was 68, 41 males (60.3%) and 27 females (39.7%). About physiological indicators results, there was no significant association between the age of the mother and sex of the baby (P=0.934). No significant association was detected between the month during which the conception occurred and the sex of the baby (P=0.734). The same result was obtained for the follicle side (P=0.236), and follicle size (P=0.659), there was no significant association between the sex of the baby with the following factors: protocol of treatment (P=0.417), IVF after HCG (P=0.237), HCG type (P=0.572), parity (P=0.282), and type of infertility (P=0.376). The cellular indicators results showed that the BMP-6 protein level in granulosa cells of mothers with daughters was almost twice as high as mothers with sons (P=0.043). But there was no significant difference between mothers with daughters and mothers with sons in PPAR? protein level (P=0.12). It can be concluded that except for BMP-6 protein level, none of the cellular and physiological indicators affects gender determination. Therefore, this cell indicator can probably be evaluated as an effective indicator in determining gender.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Granulosa Cells/metabolism , PPAR gamma/metabolism , Sex Determination Analysis/methods , Adult , Blotting, Western , Female , Fertilization in Vitro , Granulosa Cells/cytology , Humans , Infant, Newborn , Luteal Cells/cytology , Luteal Cells/metabolism , Male , Ovulation Induction/methods , Pregnancy , Young Adult
15.
Cell Mol Life Sci ; 77(18): 3627-3642, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31768607

ABSTRACT

Hypoxia-inducible factor (HIF), an αß dimer, is the master regulator of oxygen homeostasis with hundreds of hypoxia-inducible target genes. Three HIF isoforms differing in the oxygen-sensitive α subunit exist in vertebrates. While HIF-1 and HIF-2 are known transcription activators, HIF-3 has been considered a negative regulator of the hypoxia response pathway. However, the human HIF3A mRNA is subject to complex alternative splicing. It was recently shown that the long HIF-3α variants can form αß dimers that possess transactivation capacity. Here, we show that overexpression of the long HIF-3α2 variant induces the expression of a subset of genes, including the erythropoietin (EPO) gene, while simultaneous downregulation of all HIF-3α variants by siRNA targeting a shared HIF3A region leads to downregulation of EPO and additional genes. EPO mRNA and protein levels correlated with HIF3A silencing and HIF-3α2 overexpression. Chromatin immunoprecipitation analyses showed that HIF-3α2 binding associated with canonical hypoxia response elements in the promoter regions of EPO. Luciferase reporter assays showed that the identified HIF-3α2 chromatin-binding regions were sufficient to promote transcription by all three HIF-α isoforms. Based on these data, HIF-3α2 is a transcription activator that directly regulates EPO expression.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Erythropoietin/metabolism , Repressor Proteins/metabolism , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Cell Hypoxia , Cell Line, Tumor , Chromatin/metabolism , Dimerization , Erythropoietin/analysis , Erythropoietin/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA Splicing , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/metabolism , Transcriptional Activation
16.
Proc Natl Acad Sci U S A ; 115(45): 11625-11630, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30352848

ABSTRACT

Increasing age is the greatest known risk factor for the sporadic late-onset forms of neurodegenerative disorders such as Alzheimer's disease (AD). One of the brain regions most severely affected in AD is the hippocampus, a privileged structure that contains adult neural stem cells (NSCs) with neurogenic capacity. Hippocampal neurogenesis decreases during aging and the decrease is exacerbated in AD, but the mechanistic causes underlying this progressive decline remain largely unexplored. We here investigated the effect of age on NSCs and neurogenesis by analyzing the senescence accelerated mouse prone 8 (SAMP8) strain, a nontransgenic short-lived strain that spontaneously develops a pathological profile similar to that of AD and that has been employed as a model system to study the transition from healthy aging to neurodegeneration. We show that SAMP8 mice display an accelerated loss of the NSC pool that coincides with an aberrant rise in BMP6 protein, enhanced canonical BMP signaling, and increased astroglial differentiation. In vitro assays demonstrate that BMP6 severely impairs NSC expansion and promotes NSC differentiation into postmitotic astrocytes. Blocking the dysregulation of the BMP pathway and its progliogenic effect in vivo by intracranial delivery of the antagonist Noggin restores hippocampal NSC numbers, neurogenesis, and behavior in SAMP8 mice. Thus, manipulating the local microenvironment of the NSC pool counteracts hippocampal dysfunction in pathological aging. Our results shed light on interventions that may allow taking advantage of the brain's natural plastic capacity to enhance cognitive function in late adulthood and in chronic neurodegenerative diseases such as AD.


Subject(s)
Aging/genetics , Alzheimer Disease/drug therapy , Bone Morphogenetic Protein 6/genetics , Carrier Proteins/pharmacology , Neural Stem Cells/drug effects , Neuroprotective Agents/pharmacology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/metabolism , Cell Differentiation , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Injections, Intraventricular , Male , Mice , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Signal Transduction
17.
BMC Biotechnol ; 20(1): 48, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32854680

ABSTRACT

BACKGROUND: Human TGF-ß3 has been used in many studies to induce genes coding for typical cartilage matrix components and accelerate chondrogenic differentiation, making it the standard constituent in most cultivation media used for the assessment of chondrogenesis associated with various stem cell types on carrier matrices. However, in vivo data suggests that TGF-ß3 and its other isoforms also induce endochondral and intramembranous osteogenesis in non-primate species to other mammals. Based on previously demonstrated improved articular cartilage induction by a using hTGF-ß3 and hBMP-6 together on hADSC cultures and the interaction of TGF- ß with matrix in vivo, the present study investigates the interaction of a chitosan scaffold as polyanionic polysaccharide with both growth factors. The study analyzes the difference between chondrogenic differentiation that leads to stable hyaline cartilage and the endochondral ossification route that ends in hypertrophy by extending the usual panel of investigated gene expression and stringent employment of quantitative PCR. RESULTS: By assessing the viability, proliferation, matrix formation and gene expression patterns it is shown that hTGF-ß3 + hBMP-6 promotes improved hyaline articular cartilage formation in a chitosan scaffold in which ACAN with Col2A1 and not Col1A1 nor Col10A1 where highly expressed both at a transcriptional and translational level. Inversely, hTGF-ß3 alone tended towards endochondral bone formation showing according protein and gene expression patterns. CONCLUSION: These findings demonstrate that clinical therapies should consider using hTGF-ß3 + hBMP-6 in articular cartilage regeneration therapies as the synergistic interaction of these morphogens seems to ensure and maintain proper hyaline articular cartilage matrix formation counteracting degeneration to fibrous tissue or ossification. These effects are produced by interaction of the growth factors with the polysaccharide matrix.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Cartilage, Articular/metabolism , Chitosan/metabolism , Regenerative Medicine/methods , Transforming Growth Factor beta3/metabolism , Animals , Bone Morphogenetic Protein 6/genetics , Cartilage, Articular/cytology , Cell Differentiation , Cell Proliferation , Chondrogenesis/physiology , Collagen , Collagen Type X , Gene Expression , Humans , Mesenchymal Stem Cells , Osteogenesis , Stem Cells , Tissue Scaffolds , Transforming Growth Factor beta3/genetics
18.
Blood ; 132(14): 1473-1477, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30097509

ABSTRACT

Decreased hepcidin mobilizes iron, which facilitates erythropoiesis, but excess iron is pathogenic in ß-thalassemia. Erythropoietin (EPO) enhances erythroferrone (ERFE) synthesis by erythroblasts, and ERFE suppresses hepatic hepcidin production through an unknown mechanism. The BMP/SMAD pathway in the liver is critical for hepcidin control, and we show that EPO suppressed hepcidin and other BMP target genes in vivo in a partially ERFE-dependent manner. Furthermore, recombinant ERFE suppressed the hepatic BMP/SMAD pathway independently of changes in serum and liver iron. In vitro, ERFE decreased SMAD1, SMAD5, and SMAD8 phosphorylation and inhibited expression of BMP target genes. ERFE specifically abrogated the induction of hepcidin by BMP5, BMP6, and BMP7 but had little or no effect on hepcidin induction by BMP2, BMP4, BMP9, or activin B. A neutralizing anti-ERFE antibody prevented ERFE from inhibiting hepcidin induction by BMP5, BMP6, and BMP7. Cell-free homogeneous time-resolved fluorescence assays showed that BMP5, BMP6, and BMP7 competed with anti-ERFE for binding to ERFE. We conclude that ERFE suppresses hepcidin by inhibiting hepatic BMP/SMAD signaling via preferentially impairing an evolutionarily closely related BMP subgroup of BMP5, BMP6, and BMP7. ERFE can act as a natural ligand trap generated by stimulated erythropoiesis to regulate the availability of iron.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Cytokines/metabolism , Hepcidins/metabolism , Muscle Proteins/metabolism , Animals , Cell Line , Hep G2 Cells , Humans , Iron/metabolism , Liver/metabolism , Male , Mice , Signal Transduction , Smad Proteins/metabolism
19.
Blood ; 132(17): 1829-1841, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30213871

ABSTRACT

Systemic iron balance is controlled by hepcidin, a liver hormone that limits iron efflux to the bloodstream by promoting degradation of the iron exporter ferroportin in target cells. Iron-dependent hepcidin induction requires hemojuvelin (HJV), a bone morphogenetic protein (BMP) coreceptor that is disrupted in juvenile hemochromatosis, causing dramatic hepcidin deficiency and tissue iron overload. Hjv-/- mice recapitulate phenotypic hallmarks of hemochromatosis but exhibit blunted hepcidin induction following lipopolysaccharide (LPS) administration. We show that Hjv-/- mice fail to mount an appropriate hypoferremic response to acute inflammation caused by LPS, the lipopeptide FSL1, or Escherichia coli infection because residual hepcidin does not suffice to drastically decrease macrophage ferroportin levels. Hfe-/- mice, a model of milder hemochromatosis, exhibit almost wild-type inflammatory hepcidin expression and associated effects, whereas double Hjv-/-Hfe-/- mice phenocopy single Hjv-/- counterparts. In primary murine hepatocytes, Hjv deficiency does not affect interleukin-6 (IL-6)/Stat, and only slightly inhibits BMP2/Smad signaling to hepcidin; however, it severely impairs BMP6/Smad signaling and thereby abolishes synergism with the IL-6/Stat pathway. Inflammatory induction of hepcidin is suppressed in iron-deficient wild-type mice and recovers after the animals are provided overnight access to an iron-rich diet. We conclude that Hjv is required for inflammatory induction of hepcidin and controls the acute hypoferremic response by maintaining a threshold of Bmp6/Smad signaling. Our data highlight Hjv as a potential pharmacological target against anemia of inflammation.


Subject(s)
Hepatocytes/metabolism , Hepcidins/metabolism , Inflammation/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Animals , Bone Morphogenetic Protein 6/metabolism , GPI-Linked Proteins , Hemochromatosis Protein , Iron Overload/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Smad Proteins/metabolism
20.
Dev Growth Differ ; 62(2): 139-146, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32012242

ABSTRACT

Age-related macular degeneration (AMD) is one of the leading causes of blindness in elderly individuals worldwide. Oxidative stress injury to retinal pigment epithelial (RPE) cells plays a major role in the pathogenesis of AMD. The purpose of this study was to observe the correlation between Hepcidin and neovascular age-related macular degeneration (nAMD) and to further observe whether oxidative stress can inhibit Hepcidin expression through relevant signaling pathways to produce oxidative damage. We compared the concentrations of Hepcidin in the aqueous humor of nAMD patients and a control group and found that the concentration of Hepcidin was lower in nAMD patients. Through PCR and western blotting, we observed that H2 O2 can significantly inhibit the expression of Bone morphogenetic protein-6 (BMP-6) and Hepcidin and increase the intracellular iron concentration in RPE cells, while BMP-6 can reverse the inhibition of Hepcidin and the increase in iron concentration caused by H2 O2 . In addition, alterations in smad1 and smad5 expression were examined, and pretreatment with BMP-6 was demonstrated to reduce H2 O2 -induced activation of smad1 and smad5. The effects of BMP-6 were attenuated by smad1 and smad5 siRNA, further verifying that oxidative stress inhibits the expression of Hepcidin by inhibiting activation of the BMP/SMAD signaling pathway. To some extent, this study verified that oxidative stress injury plays a role in nAMD by affecting the level of hepcidin, which lays a foundation for exploring new methods to treat nAMD.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Hepcidins/metabolism , Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Aged , Blotting, Western , Bone Morphogenetic Protein 6/genetics , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Hepcidins/genetics , Humans , Macular Degeneration , Male , Middle Aged , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Smad1 Protein/genetics , Smad5 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL