Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Biol Chem ; 298(2): 101586, 2022 02.
Article in English | MEDLINE | ID: mdl-35032551

ABSTRACT

Signaling by bone morphogenetic proteins (BMPs) plays pivotal roles in embryogenesis, adult tissue homeostasis, and disease. Recent studies revealed that the well-established WNT agonist R-spondin 2 (RSPO2) is also a BMP receptor (BMP receptor type 1A) antagonist, with roles in early Xenopus embryogenesis and human acute myeloid leukemia (AML). To uncouple the BMP antagonist function from the WNT agonist function and to promote development of AML therapeutics, here we identified a 10-mer peptide (RW) derived from the thrombospondin 1 domain of RSPO2, which specifically prevents binding between RSPO2 and BMP receptor type 1A without altering WNT signaling. We also show that a corresponding RW dendrimer (RWd) exhibiting improved half-life relieves inhibition of BMP receptor signaling by RSPO2 in human AML cells, reduces cell growth, and induces differentiation. Moreover, microinjection of RWd in Xenopus embryos ventralizes the dorsoventral embryonic patterning by upregulating BMP signaling without affecting WNT signaling. Our study corroborates the function of RSPO2 as a BMP receptor antagonist and provides a proof of concept for pharmacologically uncoupling BMP antagonist from WNT agonist functions of RSPO2 using the inhibitor peptide RWd with enhanced target selectivity and limited side effects.


Subject(s)
Bone Morphogenetic Protein Receptors , Dendrimers , Leukemia, Myeloid, Acute , Wnt Proteins , Adult , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Proteins , Dendrimers/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Peptide Fragments , Proteins/pharmacology , Wnt Proteins/agonists , Wnt Signaling Pathway , Xenopus laevis
2.
Brain ; 144(8): 2291-2301, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34426831

ABSTRACT

Extrinsic inhibitors at sites of blood-brain barrier disruption and neurovascular damage contribute to remyelination failure in neurological diseases. However, therapies to overcome the extrinsic inhibition of remyelination are not widely available and the dynamics of glial progenitor niche remodelling at sites of neurovascular dysfunction are largely unknown. By integrating in vivo two-photon imaging co-registered with electron microscopy and transcriptomics in chronic neuroinflammatory lesions, we found that oligodendrocyte precursor cells clustered perivascularly at sites of limited remyelination with deposition of fibrinogen, a blood coagulation factor abundantly deposited in multiple sclerosis lesions. By developing a screen (OPC-X-screen) to identify compounds that promote remyelination in the presence of extrinsic inhibitors, we showed that known promyelinating drugs did not rescue the extrinsic inhibition of remyelination by fibrinogen. In contrast, bone morphogenetic protein type I receptor blockade rescued the inhibitory fibrinogen effects and restored a promyelinating progenitor niche by promoting myelinating oligodendrocytes, while suppressing astrocyte cell fate, with potent therapeutic effects in chronic models of multiple sclerosis. Thus, abortive oligodendrocyte precursor cell differentiation by fibrinogen is refractory to known promyelinating compounds, suggesting that blockade of the bone morphogenetic protein signalling pathway may enhance remyelinating efficacy by overcoming extrinsic inhibition in neuroinflammatory lesions with vascular damage.


Subject(s)
Blood-Brain Barrier/drug effects , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Oligodendroglia/drug effects , Remyelination/drug effects , Spinal Cord/drug effects , Animals , Blood-Brain Barrier/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Homeostasis/drug effects , Mice , Mice, Transgenic , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Spinal Cord/metabolism
3.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070207

ABSTRACT

In domestic ruminants, endometrial receptivity is related to successful pregnancy and economic efficiency. Despite several molecules having been reported in the past regarding endometrial receptivity regulation, much regarding the mechanism of endometrial receptivity regulation remains unknown due to the complex nature of the trait. In this work, we demonstrated that the cysteine-rich transmembrane bone morphogenetic protein (BMP) regulator 1 (CRIM1) served as a novel regulator in the regulation of goat endometrial receptivity in vitro. Our results showed that hormones and IFN-τ increased the expression of CRIM1 in goat endometrial epithelial cells (EECs). Knockdown of CRIM1 via specific shRNA hindered cell proliferation, cell adhesion and prostaglandins (PGs) secretion and thus derailed normal endometrial receptivity. We further confirmed that receptivity defect phenotypes due to CRIM1 interference were restored by ATG7 overexpression in EECs while a loss of ATG7 further impaired receptivity phenotypes. Moreover, our results showed that changing the expression of ATG7 affected the reactive oxygen species (ROS) production. Moreover, mR-143-5p was shown to be a potential upstream factor of CRIM1-regulated endometrial receptivity in EECs. Overall, these results suggest that CRIM1, as the downstream target of miR-143-5p, has effects on ATG7-dependent autophagy, regulating cell proliferation, cell adhesion and PG secretion, and provides a new target for the diagnosis and treatment of early pregnancy failure and for improving the success rates of artificial reproduction.


Subject(s)
Bone Morphogenetic Protein Receptors/physiology , Embryo Implantation/genetics , Endometrium/physiology , Goats/physiology , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy/physiology , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/physiology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/genetics , Cell Adhesion , Cell Proliferation , Cells, Cultured , Embryo Implantation/physiology , Endometrium/cytology , Endometrium/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Estradiol/pharmacology , Female , Gene Knockdown Techniques , Goats/genetics , Interferon Type I/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Pregnancy , Pregnancy Proteins/pharmacology , Progesterone/pharmacology , Prostaglandins/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Up-Regulation
4.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670533

ABSTRACT

Hereditary hemorrhagic telangiectasia type 1 (HHT1) is a severe vascular disorder caused by mutations in the TGFß/BMP co-receptor endoglin. Endoglin haploinsufficiency results in vascular malformations and impaired neoangiogenesis. Furthermore, HHT1 patients display an impaired immune response. To date it is not fully understood how endoglin haploinsufficient immune cells contribute to HHT1 pathology. Therefore, we investigated the immune response during tissue repair in Eng+/- mice, a model for HHT1. Eng+/- mice exhibited prolonged infiltration of macrophages after experimentally induced myocardial infarction. Moreover, there was an increased number of inflammatory M1-like macrophages (Ly6Chigh/CD206-) at the expense of reparative M2-like macrophages (Ly6Clow/CD206+). Interestingly, HHT1 patients also showed an increased number of inflammatory macrophages. In vitro analysis revealed that TGFß-induced differentiation of Eng+/- monocytes into M2-like macrophages was blunted. Inhibiting BMP signaling by treating monocytes with LDN-193189 normalized their differentiation. Finally, LDN treatment improved heart function after MI and enhanced vascularization in both wild type and Eng+/- mice. The beneficial effect of LDN was also observed in the hind limb ischemia model. While blood flow recovery was hampered in vehicle-treated animals, LDN treatment improved tissue perfusion recovery in Eng+/- mice. In conclusion, BMPR kinase inhibition restored HHT1 macrophage imbalance in vitro and improved tissue repair after ischemic injury in Eng+/- mice.


Subject(s)
Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Disease Models, Animal , Endoglin/metabolism , Myocardial Infarction/prevention & control , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Wound Healing/drug effects , Animals , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Cells, Cultured , Endoglin/genetics , Female , Heterozygote , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/immunology , Telangiectasia, Hereditary Hemorrhagic/metabolism , Wound Healing/genetics
5.
Nat Commun ; 10(1): 4533, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586071

ABSTRACT

Multiple myeloma is an incurable, bone marrow-dwelling malignancy that disrupts bone homeostasis causing skeletal damage and pain. Mechanisms underlying myeloma-induced bone destruction are poorly understood and current therapies do not restore lost bone mass. Using transcriptomic profiling of isolated bone lining cell subtypes from a murine myeloma model, we find that bone morphogenetic protein (BMP) signalling is upregulated in stromal progenitor cells. BMP signalling has not previously been reported to be dysregulated in myeloma bone disease. Inhibition of BMP signalling in vivo using either a small molecule BMP receptor antagonist or a solubilized BMPR1a-FC receptor ligand trap prevents trabecular and cortical bone volume loss caused by myeloma, without increasing tumour burden. BMP inhibition directly reduces osteoclastogenesis, increases osteoblasts and bone formation, and suppresses bone marrow sclerostin levels. In summary we describe a novel role for the BMP pathway in myeloma-induced bone disease that can be therapeutically targeted.


Subject(s)
Bone Diseases/drug therapy , Bone Morphogenetic Proteins/metabolism , Multiple Myeloma/complications , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Stem Cells/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Density/drug effects , Bone Diseases/etiology , Bone Diseases/pathology , Bone Marrow/pathology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Cell Line, Tumor , Disease Models, Animal , Femur/cytology , Femur/drug effects , Femur/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred Strains , Multiple Myeloma/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , RNA-Seq , Signal Transduction/drug effects , Stem Cells/pathology , Tibia/cytology , Tibia/drug effects , Tibia/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Metab Eng ; 52: 57-67, 2019 03.
Article in English | MEDLINE | ID: mdl-30447330

ABSTRACT

A Chinese hamster ovary (CHO) cell line producing recombinant human bone morphogenetic protein-4 (rhBMP-4) (CHO-BMP-4), which expresses essential components of BMP signal transduction, underwent autocrine BMP-4 signaling. RNA seq analysis on CHO host cells (DG44) treated with rhBMP-4 (20 µg/mL) suggested that rhBMP-4 induced signaling in CHO cells could be a critical factor in limiting rhBMP-4 production and should be removed to improve rhBMP-4 production in recombinant CHO (rCHO) cells. The inhibition of autocrine BMP signaling in CHO-BMP-4 cells by the addition of LDN-193189, a chemical inhibitor of BMP receptor type I, significantly increased the mRNA expression levels of rhBMP-4. To establish BMP signaling-free host cells, a BMP receptor, the BMPRIA or BMPRII gene in DG44 cells, was knocked out using CRISPR/Cas9 gene-editing technology. Using three different knockout (KO) host cell lines as well as a DG44 wild-type (wt) cell line, rCHO cell clones producing rhBMP-4 were generated by a stepwise selection with increasing methotrexate concentrations. KO-derived clones showed a significantly higher maximum rhBMP-4 concentration than wt-derived clones in both batch and fed-batch cultures. Unlike wt-derived clones, KO-derived cell clones were able to produce higher amounts of hBMP-4 transcripts and proteins in the stationary phase of growth and did not experience growth inhibition induced by rhBMP-4. The mean maximum rhBMP-4 concentration of KO host-derived clones was approximately 2.4-fold higher than that of wt-derived clones (P < 0.05). Taken together, the disruption of BMP signaling in CHO cells by knocking out the BMP receptor significantly improved rhBMP-4 production.


Subject(s)
Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors/genetics , Animals , Antimetabolites/pharmacology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , CHO Cells , CRISPR-Cas Systems , Cricetinae , Cricetulus , Feedback, Physiological , Gene Expression Profiling , Gene Knockout Techniques , Homeostasis , Methotrexate/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Recombinant Proteins
7.
Oncogene ; 37(27): 3672-3685, 2018 07.
Article in English | MEDLINE | ID: mdl-29622797

ABSTRACT

BMP receptor inhibitors induce death of cancer cells through the downregulation of antiapoptotic proteins XIAP, pTAK1, and Id1-Id3. However, the current most potent BMP receptor inhibitor, DMH2, does not downregulate BMP signaling in vivo because of metabolic instability and poor pharmacokinetics. Here we identified the site of metabolic instability of DMH2 and designed a novel BMP receptor inhibitor, JL5. We show that JL5 has a greater volume of distribution and suppresses the expression of Id1 and pTak1 in tumor xenografts. Moreover, we demonstrate JL5-induced tumor cell death and tumor regression in xenograft mouse models without immune cells and humanized with adoptively transferred human immune cells. In humanized mice, JL5 additionally induces the infiltration of immune cells within the tumor microenvironment. Our studies show that the BMP signaling pathway is targetable in vivo and BMP receptor inhibitors can be developed as a therapeutic to treat cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolones/pharmacology , Tumor Microenvironment/drug effects , A549 Cells , Adoptive Transfer , Animals , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Quinolones/chemistry , Quinolones/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Hepatology ; 68(1): 274-288, 2018 07.
Article in English | MEDLINE | ID: mdl-29315687

ABSTRACT

During development, the endoderm initiates organ-restricted gene expression patterns in a spatiotemporally controlled manner. This process, termed induction, requires signals from adjacent mesodermal derivatives. Fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) emanating from the cardiac mesoderm and the septum transversum mesenchyme (STM), respectively, are believed to be simultaneously and uniformly required to directly induce hepatic gene expression from the murine endoderm. Using small molecule inhibitors of BMP signals during liver bud induction in the developing mouse embryo, we found that BMP signaling was not uniformly required to induce hepatic gene expression. Although BMP inhibition caused an overall reduction in the number of induced hepatoblasts, the STM-bounded posterior liver bud demonstrated the most severe loss of the essential hepatic transcription factor, hepatocyte nuclear factor 4-α (HNF4α), whereas the sinus venosus-lined anterior liver bud was less affected. We found that the posterior liver bud progenitors were anteriorly displaced and aberrantly activated pancreatobiliary markers, including sex-determining region Y-box 9 (SOX9). Additionally, we found that ectopically expressed SOX9 inhibited HNF4α and that BMP was indirectly required for hepatoblast induction. Finally, because previous studies have demonstrated that FGF signals are essential for anterior but not posterior liver bud induction, we examined synchronous BMP and FGF inhibition and found this led to a nearly complete loss of hepatoblasts. CONCLUSION: BMP signaling is required to maintain the hepato-pancreatobiliary boundary, at least in part, by indirectly repressing SOX9 in the hepatic endoderm. BMP and FGF signals are each required for the induction of spatially complementary subsets of hepatoblasts. These results underscore the importance of studying early inductive processes in the whole embryo. (Hepatology 2018;68:274-288).


Subject(s)
Bone Morphogenetic Proteins/physiology , Embryonic Induction , Liver/embryology , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Cell Proliferation , Fibroblast Growth Factors/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Mice , SOX9 Transcription Factor/metabolism
9.
Bone ; 109: 251-258, 2018 04.
Article in English | MEDLINE | ID: mdl-28918311

ABSTRACT

Individuals with the rare developmental disorder fibrodysplasia ossificans progressiva (FOP) experience disabling heterotopic ossification caused by a gain of function mutation in the intracellular region of the BMP type I receptor kinase ALK2, encoded by the gene ACVR1. Small molecule BMP type I receptor inhibitors that block this ossification in FOP mouse models have been derived from the pyrazolo[1,5-a]pyrimidine scaffold of dorsomorphin. While the first derivative LDN-193189 exhibited pan inhibition of BMP receptors, the more recent compound LDN-212854 has shown increased selectivity for ALK2. Here we solved the crystal structure of ALK2 in complex with LDN-212854 to define how its binding interactions compare to previously reported BMP and TGFß receptor inhibitors. LDN-212854 bound to the kinase hinge region as a typical type I ATP-competitive inhibitor with a single hydrogen bond to ALK2 His286. Specificity arising from the 5-quinoline moiety was associated with a distinct pattern of water-mediated hydrogen bonds involving Lys235 and Glu248 in the inactive conformation favoured by ALK2. The structure of this complex provides a template for the design of future ALK2 inhibitors under development for the treatment of FOP and other related conditions of heterotopic ossification.


Subject(s)
Activin Receptors, Type I/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Quinolines/metabolism , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Myositis Ossificans/metabolism , Protein Binding , Pyrazoles/chemistry , Pyrimidines/chemistry , Quinolines/chemistry , Sf9 Cells
10.
Oncotarget ; 8(6): 10145-10160, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28052036

ABSTRACT

Angiomotin (AMOT) is a family of proteins found to be a component of the apical junctional complex of vertebrate epithelial cells and is recently found to play important roles in neurofibromatosis type 2 (NF-2). Whether AMOT plays a role in prostate cancer (PCa) is unknown. AMOT is expressed as two isoforms, AMOTp80 and AMOTp130, which has a 409 aa N-terminal domain that is absent in AMOTp80. Both AMOTp80 and AMOTp130 are expressed in LNCaP and C4-2B4, but at a low to undetectable level in PC3, DU145, and BPH1 cells. Further study showed that AMOTp130 and AMOTp80 have distinct functions in PCa cells. We found that AMOTp80, but not AMOT p130, functioned as a tumor promoter by enhancing PCa cell proliferation. Mechanistic studies showed that AMOTp80 signaled through the Hippo pathway by promoting nuclear translocation of YAP, resulting in an increased expression of YAP target protein BMP4. Moreover, inhibition of BMP receptor activity by LDN-193189 abrogates AMOTp80-mediated cell proliferation. Together, this study reveals a novel mechanism whereby the AMOTp80-Merlin-MST1-LATS-YAP-BMP4 pathway leads to AMOTp80-induced tumor cell proliferation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/genetics , Angiomotins , Animals , Antineoplastic Agents/pharmacology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice, SCID , Microfilament Proteins , Phosphoproteins/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transcription Factors , Transfection , YAP-Signaling Proteins
11.
Nat Commun ; 7: 13796, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27982029

ABSTRACT

Some polypeptide N-acetyl-galactosaminyltransferases (GALNTs) are associated with cancer, but their function in organ-specific metastasis remains unclear. Here, we report that GALNT14 promotes breast cancer metastasis to the lung by enhancing the initiation of metastatic colonies as well as their subsequent growth into overt metastases. Our results suggest that GALNT14 augments the self-renewal properties of breast cancer cells (BCCs). Furthermore, GALNT14 overcomes the inhibitory effect of lung-derived bone morphogenetic proteins (BMPs) on self-renewal and therefore facilitates metastasis initiation within the lung microenvironment. In addition, GALNT14 supports continuous growth of BCCs in the lung by not only inducing macrophage infiltration but also exploiting macrophage-derived fibroblast growth factors (FGFs). Finally, we identify KRAS-PI3K-c-JUN signalling as an upstream pathway that accounts for the elevated expression of GALNT14 in lung-metastatic BCCs. Collectively, our findings uncover an unprecedented role for GALNT14 in the pulmonary metastasis of breast cancer and elucidate the underlying molecular mechanisms.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/secondary , N-Acetylgalactosaminyltransferases/metabolism , Animals , Benzamides/pharmacology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Breast Neoplasms/metabolism , Cell Line, Tumor , Diamines/pharmacology , Dioxoles/pharmacology , Female , Glycosylation , Guanine Nucleotide Exchange Factors , Humans , Lung/pathology , Lung/physiology , Mice , Mice, Nude , Mutation , N-Acetylgalactosaminyltransferases/genetics , Neoplasms, Experimental/pathology , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptors, Notch/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Thiazoles/pharmacology , Polypeptide N-acetylgalactosaminyltransferase
12.
Oncotarget ; 7(12): 14951-62, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26908452

ABSTRACT

BACKGROUND & AIMS: Breast cancer (BC) is prevalent worldwide malignant cancer. Improvements in timely and effective diagnosis and prediction are needed. As reported, secreted DAND5 is contributed to BC metastasis. We aim to assess whether DAND5 in peripheral blood serum could determine BC-specific mortality. METHODS: We used immunohistochemistry staining to detect DAND5 expression in our BC tissue array including 250 samples. Angiogenesis assay and xenograft mice model were used to examine the secreted DAND5 function in BC progression. Serum concentration of DAND5 was examined by ELISA in 1730 BC patients. Kaplan-Meier and adjusted Cox proportional hazards models were utilized to analyze the prognosis and survival of BC patients. RESULTS: Tissue array results showed that positive DAND5 staining cases displayed a higher likelihood of occurrence of disease events (HR=5.494; 95% CI: 1.008-2.353; P=0.048) in univariate analysis and remained the same trend in multivariate analysis (HR=2.537; 95% CI: 1.056-6.096; P=0.037). DAND5 positive patients exerted generally poor DFS (P=0.041) in the Kaplan-Meier survival analysis. Furthermore, secreted DAND5 promoted tumor growth and angiogenesis in vitro and in vivo. In addition, positive DAND5 in BC patients serum was associated with increased risk of disease events occurrence (univariate: HR=1.58; 95% CI: 1.206-2.070; P=0.001; multivariate: HR=1.4; 95% CI: 1.003-1.954; P=0.048) in univariate and multivariate survival analysis. In the Kaplan-Meier analysis, serum DAND5 positively correlated with poor DFS (P=0.001) and DDFS (P=0.002). CONCLUSIONS: DAND5 was correlated with poor survival and could serve as an easily detectable serum biomarker to predict the survival of breast cancer.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/mortality , Intercellular Signaling Peptides and Proteins/blood , Neoplasm Recurrence, Local/mortality , Neovascularization, Pathologic/pathology , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Female , Follow-Up Studies , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Neovascularization, Pathologic/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Expert Rev Hematol ; 9(2): 169-86, 2016.
Article in English | MEDLINE | ID: mdl-26669208

ABSTRACT

The secreted peptide hormone hepcidin regulates systemic and local iron homeostasis through degradation of the iron exporter ferroportin. Dysregulation of hepcidin leads to altered iron homeostasis and development of pathological disorders including hemochromatosis, and iron loading and iron restrictive anemias. Therapeutic modulation of hepcidin is a promising method to ameliorate these conditions. Several approaches have been taken to enhance or reduce the effects of hepcidin in vitro and in vivo. Based on these approaches, hepcidin modulating drugs have been developed and are undergoing clinical evaluation. In this article we review the rationale for development of these drugs, the data concerning their safety and efficacy, their therapeutic uses, and potential future prospects.


Subject(s)
Hepcidins/metabolism , Iron Metabolism Disorders/metabolism , Iron Metabolism Disorders/therapy , Iron/metabolism , Animals , Biological Transport , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Cation Transport Proteins/metabolism , Gene Expression Regulation , Hepcidins/agonists , Hepcidins/antagonists & inhibitors , Hepcidins/deficiency , Homeostasis , Humans , Interleukin-6/metabolism , Iron Metabolism Disorders/genetics , Peptide Hormones/pharmacology , Peptide Hormones/therapeutic use , Signal Transduction/drug effects
14.
Biochem Soc Trans ; 43(5): 795-800, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26517884

ABSTRACT

Chordin-mediated regulation of bone morphogenetic protein (BMP) family growth factors is essential in early embryogenesis and adult homoeostasis. Chordin binds to BMPs through cysteine-rich von Willebrand factor type C (vWC) homology domains and blocks them from interacting with their cell surface receptors. These domains also self-associate and enable chordin to target related proteins to fine-tune BMP regulation. The chordin-BMP inhibitory complex is strengthened by the secreted glycoprotein twisted gastrulation (Tsg); however, inhibition is relieved by cleavage of chordin at two specific sites by tolloid family metalloproteases. As Tsg enhances this cleavage process, it serves a dual role as both promoter and inhibitor of BMP signalling. Recent developments in chordin research suggest that rather than simply being by-products, the cleavage fragments of chordin continue to play a role in BMP regulation. In particular, chordin cleavage at the C-terminus potentiates its anti-BMP activity in a type-specific manner.


Subject(s)
Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Proteins/antagonists & inhibitors , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Models, Biological , Proteins/metabolism , Signal Transduction , Tolloid-Like Metalloproteinases/metabolism , Animals , Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Protein Receptors/chemistry , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/metabolism , Glycoproteins/chemistry , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Stability , Proteins/chemistry , Proteolysis , Tolloid-Like Metalloproteinases/chemistry
15.
Molecules ; 20(5): 7586-601, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25919279

ABSTRACT

Bone morphogenetic proteins (BMPs) control many developmental and physiological processes, including skeleton formation and homeostasis. Previous studies in zebrafish revealed the crucial importance of proper BMP signaling before 48 h post-fertilization (hpf) for cartilage formation in the skull. Here, we focus on the involvement of the BMP pathway between 48 and 96 hpf in bone formation after 96 hpf. Using BMP inhibitors and the expression of a dominant-negative BMP receptor, we analyze whether the loss of BMP signaling affects osteoblastogenesis, osteoblast function and bone mineralization. To this end, we used the transgenic zebrafish line Tg(osterix:mCherry), detection of nitric oxide (NO) production, and alizarin red staining, respectively. We observed that inhibition of BMP signaling between 48 and 72 hpf led to a reduction of NO production and bone mineralization. Osteoblast maturation and chondrogenesis, on the other hand, seemed unchanged. Osteoblast function and bone formation were less affected when BMP signaling was inhibited between 72 and 96 hpf. These results suggest that for the onset of bone formation, proper BMP signaling between 48 and 72 hpf is crucial to ensure osteoblast function and ossification. Furthermore, detection of NO in developing zebrafish larvae appears as an early indicator of bone calcification activity.


Subject(s)
Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Proteins/antagonists & inhibitors , Calcification, Physiologic/physiology , Nitric Oxide/biosynthesis , Osteogenesis/physiology , AMP-Activated Protein Kinases/antagonists & inhibitors , Aminopyridines/pharmacology , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein Receptors/biosynthesis , Bone Morphogenetic Proteins/metabolism , Chondrogenesis/physiology , Osteoblasts/metabolism , Phenols/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Skull/cytology , Skull/embryology , Skull/metabolism , Zebrafish
16.
Curr Cancer Drug Targets ; 14(7): 652-8, 2014.
Article in English | MEDLINE | ID: mdl-25088037

ABSTRACT

Cysteine-rich motor neuron1 protein (CRIM1), a novel antagonist of bone morphogenetic proteins (BMPs), is reported to regulate the processing of BMPs preprotein into mature protein and the delivery of BMPs to the cell surface. Previous studies have shown that CRIM1 is an important player in regulating placental development, organogenesis, angiogenesis and kidney disease. Here, we propose that CRIM1 is a potential risk factor in cancer progression and metastasis. The epithelial-mesenchymal transition (EMT), which is characterized by the loss of epithelial phenotype and the acquisition of mesenchymal characteristics, is closely associated with invasion and metastasis of tumors. At the same time, it is hard for us to ignore the importance of angiogenesis in the genesis and progression of cancer. In this review we summarized the construction and previous researches of CRIM1. Furthermore, as it may be involved in tumor development and progression through its potential role in the EMT, capillary formation and angiogenesis maintenance, we proposed for the first time that CRIM1 may be a cancer related factor.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Models, Biological , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Disease Progression , Epithelial-Mesenchymal Transition , Humans , Neoplasm Metastasis , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/epidemiology , Neoplasms/pathology , Neoplasms/physiopathology , Neovascularization, Pathologic/etiology , Risk Factors
17.
Blood ; 124(3): 393-402, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24894772

ABSTRACT

Bone morphogenetic protein (BMP) signaling regulates early hematopoietic development, proceeding from mesoderm patterning through the progressive commitment and differentiation of progenitor cells. The BMP pathway signals largely through receptor-mediated activation of Mothers Against Decapentaplegic homolog (SMAD) proteins, although alternate pathways are modulated through various components of mitogen-activated protein kinase (MAPK) signaling. Using a conditional, short hairpin RNA (shRNA)-based knockdown system in the context of differentiating embryonic stem cells (ESCs), we demonstrated previously that Smad1 promotes hemangioblast specification, but then subsequently restricts primitive progenitor potential. Here we show that co-knockdown of Smad5 restores normal progenitor potential of Smad1-depleted cells, suggesting opposing functions for Smad1 and Smad5. This balance was confirmed by cotargeting Smad1/5 with a specific chemical antagonist, LDN193189 (LDN). However, we discovered that LDN treatment after hemangioblast commitment enhanced primitive myeloid potential. Moreover, inhibition with LDN (but not SMAD depletion) increased expression of Delta-like ligands Dll1 and Dll3 and NOTCH activity; abrogation of NOTCH activity restored LDN-enhanced myeloid potential back to normal, corresponding with expression levels of the myeloid master regulator, C/EBPα. LDN but not SMAD activity was also associated with activation of the p38MAPK pathway, and blocking this pathway was sufficient to enhance myelopoiesis. Therefore, NOTCH and p38MAPK pathways balance primitive myeloid progenitor output downstream of the BMP pathway.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Myelopoiesis/physiology , Receptors, Notch/metabolism , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Calcium-Binding Proteins , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Myelopoiesis/drug effects , Myelopoiesis/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/genetics , Smad5 Protein/metabolism
18.
Development ; 141(2): 448-59, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353059

ABSTRACT

Angiogenesis defines the process in which new vessels grow from existing vessels. Using the mouse retina as a model system, we show that cysteine-rich motor neuron 1 (Crim1), a type I transmembrane protein, is highly expressed in angiogenic endothelial cells. Conditional deletion of the Crim1 gene in vascular endothelial cells (VECs) causes delayed vessel expansion and reduced vessel density. Based on known Vegfa binding by Crim1 and Crim1 expression in retinal vasculature, where angiogenesis is known to be Vegfa dependent, we tested the hypothesis that Crim1 is involved in the regulation of Vegfa signaling. Consistent with this hypothesis, we showed that VEC-specific conditional compound heterozygotes for Crim1 and Vegfa exhibit a phenotype that is more severe than each single heterozygote and indistinguishable from that of the conditional homozygotes. We further showed that human CRIM1 knockdown in cultured VECs results in diminished phosphorylation of VEGFR2, but only when VECs are required to rely on an autocrine source of VEGFA. The effect of CRIM1 knockdown on reducing VEGFR2 phosphorylation was enhanced when VEGFA was also knocked down. Finally, an anti-VEGFA antibody did not enhance the effect of CRIM1 knockdown in reducing VEGFR2 phosphorylation caused by autocrine signaling, but VEGFR2 phosphorylation was completely suppressed by SU5416, a small-molecule VEGFR2 kinase inhibitor. These data are consistent with a model in which Crim1 enhances the autocrine signaling activity of Vegfa in VECs at least in part via Vegfr2.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Retinal Vessels/growth & development , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor A/metabolism , Alleles , Animals , Autocrine Communication , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/genetics , Cell Proliferation , Endothelial Cells/metabolism , Gene Knockdown Techniques , Heterozygote , Homozygote , Human Umbilical Vein Endothelial Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Mice, Transgenic , Neovascularization, Physiologic , Pericytes/metabolism , Phenotype , Phosphorylation , RNA, Small Interfering/genetics , Retinal Vessels/embryology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Bioorg Med Chem Lett ; 23(11): 3248-52, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23639540

ABSTRACT

A structure-activity relationship of the 3- and 6-positions of the pyrazolo[1,5-a]pyrimidine scaffold of the known BMP inhibitors dorsomorphin, 1, LDN-193189, 2, and DMH1, 3, led to the identification of a potent and selective compound for ALK2 versus ALK3. The potency contributions of several 3-position substituents were evaluated with subtle structural changes leading to significant changes in potency. From these studies, a novel 5-quinoline molecule was identified and designated an MLPCN probe molecule, ML347, which shows >300-fold selectivity for ALK2 and presents the community with a selective molecular probe for further biological evaluation.


Subject(s)
Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemistry , Quinolines/chemistry , Quinolines/chemical synthesis , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein Receptors/metabolism , Half-Life , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Humans , Mice , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Quinolines/metabolism , Quinolines/pharmacokinetics , Rats , Structure-Activity Relationship
20.
PLoS One ; 8(4): e59442, 2013.
Article in English | MEDLINE | ID: mdl-23560048

ABSTRACT

Bone Morphogenetic Proteins (BMPs) are important growth factors that regulate many cellular processes. During embryogenesis they act as morphogens and play a critical role during organ development. They influence cell fates via concentration-gradients in the embryos where cells transduce this extracellular information into gene expression profiles and cell fate decisions. How receiving cells decode and quantify BMP2/4 signals is hardly understood. There is little data on the quantitative relationships between signal input, transducing molecules, their states and location, and ultimately their ability to integrate graded systemic inputs and generate qualitative responses. Understanding this signaling network on a quantitative level should be considered a prerequisite for efficient pathway modulation, as the BMP pathway is a prime target for therapeutic invention. Hence, we quantified the spatial distribution of the main signal transducer of the BMP2/4 pathway in response to different types and levels of stimuli in c2c12 cells. We found that the subcellular localization of Smad1 is independent of ligand concentration. In contrast, Smad1 phosphorylation levels relate proportionally to BMP2 ligand concentrations and they are entirely located in the nucleus. Interestingly, we found that BMP2 stimulates target gene expression in non-linear, wave-like forms. Amplitudes showed a clear concentration-dependency, for sustained and transient stimulation. We found that even burst-stimulation triggers gene-expression wave-like modulations that are detectable for at least 30 h. Finally, we show here that target gene expression oscillations depend on receptor kinase activity, as the kinase drives further expression pulses without receptor reactivation and the target gene expression breaks off after inhibitor treatment in c2c12 cells.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors/genetics , Myoblasts/metabolism , Smad1 Protein/genetics , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter , Luciferases , Mice , Myoblasts/cytology , Myoblasts/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Smad1 Protein/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...