Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
BMJ Open Respir Res ; 10(1)2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38568728

ABSTRACT

BACKGROUND: Collagen XIII is a transmembrane collagen associated with neuromuscular junction development, and in humans its deficiency results in congenital myasthenic syndrome type 19 (CMS19), which leads to breathing difficulties. CMS19 patients usually have restricted lung capacity and one patient developed chronic lung disease. In single-cell RNA sequencing studies, collagen XIII has been identified as a marker for pulmonary lipofibroblasts, which have been implicated in the resolution of pulmonary fibrosis. METHODS: We investigated the location and function of collagen XIII in the lung to understand the origin of pulmonary symptoms in human CMS19 patients. Additionally, we performed immunostainings on idiopathic pulmonary fibrosis (IPF) samples (N=5) and both normal and fibrotic mouse lung. To study whether the lack of collagen XIII predisposes to restrictive lung disease, we exposed Col13a1-modified mice to bleomycin-induced pulmonary fibrosis. RESULTS: Apparently normal alveolar septum sections of IPF patients' lungs stained faintly for collagen XIII, and its expression was pinpointed to the septal fibroblasts in the mouse lung. Lung capacity was increased in mice lacking collagen XIII by over 10%. In IPF samples, collagen XIII was expressed by basal epithelial cells, hyperplastic alveolar epithelial cells and stromal cells in fibrotic areas, but the development of pulmonary fibrosis was unaffected in collagen XIII-deficient mice. CONCLUSIONS: Changes in mouse lung function appear to represent a myasthenic manifestation of collagen XIII deficiency. We suggest that respiratory muscle myasthenia is the primary cause of the breathing problems suffered by CMS19 patients in addition to skeletal deformities. Induction of collagen XIII expression in the IPF patients' lungs warrants further studies to reveal collagen XIII-dependent disease mechanisms.


Subject(s)
Idiopathic Pulmonary Fibrosis , Respiratory Physiological Phenomena , Humans , Animals , Mice , Dyspnea , Collagen , Lung
2.
PLoS One ; 17(10): e0276215, 2022.
Article in English | MEDLINE | ID: mdl-36251700

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a severe lung disease with a poor prognosis and few treatment options. In the most widely used experimental model for this disease, bleomycin is administered into the lungs of mice, causing a reaction of inflammation and consequent fibrosis that resembles the progression of human IPF. The inflammation and fibrosis together induce changes in gene expression that can be analyzed with reverse transcription quantitative real-time PCR (RT-qPCR), in which accurate normalization with a set of stably expressed reference genes is critical for obtaining reliable results. This work compares ten commonly used candidate reference genes in the late, fibrotic phase of bleomycin-induced pulmonary fibrosis and ranks them from the most to the least stable using NormFinder and geNorm. Sdha, Polr2a and Hprt were identified as the best performing and least variable reference genes when alternating between normal and fibrotic conditions. In order to validate the findings, we investigated the expression of Tnf and Col1a1, representing the hallmarks of inflammation and fibrotic changes, respectively. With the best three genes as references, both were found to be upregulated relative to untreated controls, unlike the situation when analyzed solely with Gapdh, a commonly used reference gene. We therefore recommend Sdha, Polr2a and Hprt as reference genes for RT-qPCR in the 4-week bleomycin challenge that represents the late fibrotic phase.


Subject(s)
Bleomycin , Pulmonary Fibrosis , Animals , Gene Expression Profiling/methods , Humans , Hypoxanthine Phosphoribosyltransferase , Inflammation , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Real-Time Polymerase Chain Reaction/methods , Reference Standards , Reverse Transcription
3.
Front Oncol ; 12: 981009, 2022.
Article in English | MEDLINE | ID: mdl-36003785

ABSTRACT

Integrin α11ß1 is a collagen-binding integrin that is needed to induce and maintain the myofibroblast phenotype in fibrotic tissues and during wound healing. The expression of the α11 is upregulated in cancer-associated fibroblasts (CAFs) in various human neoplasms. We investigated α11 expression in human cutaneous squamous cell carcinoma (cSCC) and in benign and premalignant human skin lesions and monitored its effects on cSCC development by subjecting α11-knockout (Itga11-/- ) mice to the DMBA/TPA skin carcinogenesis protocol. α11-deficient mice showed significantly decreased tumor cell proliferation, leading to delayed tumor development and reduced tumor burden. Integrin α11 expression was significantly upregulated in the desmoplastic tumor stroma of human and mouse cSCCs, and the highest α11 expression was detected in high-grade tumors. Our results point to a reduced ability of α11-deficient stromal cells to differentiate into matrix-producing and tumor-promoting CAFs and suggest that this is one causative mechanism underlying the observed decreased tumor growth. An unexpected finding in our study was that, despite reduced CAF activation, the α11-deficient skin tumors were characterized by the presence of thick and regularly aligned collagen bundles. This finding was attributed to a higher expression of TGFß1 and collagen crosslinking lysyl oxidases in the Itga11-/- tumor stroma. In summary, our data suggest that α11ß1 operates in a complex interactive tumor environment to regulate ECM synthesis and collagen organization and thus foster cSCC growth. Further studies with advanced experimental models are still needed to define the exact roles and molecular mechanisms of stromal α11ß1 in skin tumorigenesis.

4.
J Biol Chem ; 298(8): 102222, 2022 08.
Article in English | MEDLINE | ID: mdl-35787374

ABSTRACT

Hypoxia-inducible factor (HIF) prolyl 4-hydroxylases (HIF-P4Hs 1-3) are druggable targets in renal anemia, where pan-HIF-P4H inhibitors induce an erythropoietic response. Preclinical data suggest that HIF-P4Hs could also be therapeutic targets for treating metabolic dysfunction, although the contributions of HIF-P4H isoenzymes in various tissues to the metabolic phenotype are inadequately understood. Here, we used mouse lines that were gene-deficient for HIF-P4Hs 1 to 3 and two preclinical pan-HIF-P4H inhibitors to study the contributions of these isoenzymes to the anthropometric and metabolic outcome and HIF response. We show both inhibitors induced a HIF response in wildtype white adipose tissue (WAT), liver, and skeletal muscle and alleviated metabolic dysfunction during a 6-week treatment period, but they did not alter healthy metabolism. Our data indicate that HIF-P4H-1 contributed especially to skeletal muscle and WAT metabolism and that its loss lowered body weight and serum cholesterol levels upon aging. In addition, we found HIF-P4H-3 had effects on the liver and WAT and its loss increased body weight, adiposity, liver weight and triglyceride levels, WAT inflammation, and cholesterol levels and resulted in hyperglycemia and insulin resistance, especially during aging. Finally, we demonstrate HIF-P4H-2 affected all tissues studied; its inhibition lowered body and liver weight and serum cholesterol levels and improved glucose tolerance. We found very few HIF target metabolic mRNAs were regulated by the inhibition of three isoenzymes, thus suggesting a potential for selective therapeutic tractability. Altogether, these data provide specifications for the future development of HIF-P4H inhibitors for the treatment of metabolic diseases.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Isoenzymes , Adipose Tissue, White/metabolism , Aging/metabolism , Animals , Body Weight , Cholesterol/blood , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Insulin Resistance , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Liver/metabolism , Mice , Muscle, Skeletal/metabolism , Obesity/metabolism
5.
J Biol Chem ; 298(4): 101787, 2022 04.
Article in English | MEDLINE | ID: mdl-35247391

ABSTRACT

Hypoxia-inducible factors (HIFs) induce numerous genes regulating oxygen homeostasis. As oxygen sensors of the cells, the HIF prolyl 4-hydroxylases (HIF-P4Hs) regulate the stability of HIFs in an oxygen-dependent manner. During hair follicle (HF) morphogenesis and cycling, the location of dermal papilla (DP) alternates between the dermis and hypodermis and results in varying oxygen levels for the DP cells. These cells are known to express hypoxia-inducible genes, but the role of the hypoxia response pathway in HF development and homeostasis has not been studied. Using conditional gene targeting and analysis of hair morphogenesis, we show here that lack of Hif-p4h-2 in Forkhead box D1 (FoxD1)-lineage mesodermal cells interferes with the normal HF development in mice. FoxD1-lineage cells were found to be mainly mesenchymal cells located in the dermis of truncal skin, including those cells composing the DP of HFs. We found that upon Hif-p4h-2 inactivation, HF development was disturbed during the first catagen leading to formation of epithelial-lined HF cysts filled by unorganized keratins, which eventually manifested as truncal alopecia. Furthermore, the depletion of Hif-p4h-2 led to HIF stabilization and dysregulation of multiple genes involved in keratin formation, HF differentiation, and HIF, transforming growth factor ß (TGF-ß), and Notch signaling. We hypothesize that the failure of HF cycling is likely to be mechanistically caused by disruption of the interplay of the HIF, TGF-ß, and Notch pathways. In summary, we show here for the first time that HIF-P4H-2 function in FoxD1-lineage cells is essential for the normal development and homeostasis of HFs.


Subject(s)
Alopecia , Hypoxia-Inducible Factor-Proline Dioxygenases , Alopecia/enzymology , Alopecia/genetics , Animals , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice , Oxygen/metabolism , Transforming Growth Factor beta
6.
Angiogenesis ; 25(2): 259-274, 2022 05.
Article in English | MEDLINE | ID: mdl-34997404

ABSTRACT

Hypoxia plays an important regulatory role in the vasculature to adjust blood flow to meet metabolic requirements. At the level of gene transcription, the responses are mediated by hypoxia-inducible factor (HIF) the stability of which is controlled by the HIF prolyl 4-hydroxylase-2 (PHD2). In the lungs hypoxia results in vasoconstriction, however, the pathophysiological relevance of PHD2 in the major arterial cell types; endothelial cells (ECs) and arterial smooth muscle cells (aSMCs) in the adult vasculature is incompletely characterized. Here, we investigated PHD2-dependent vascular homeostasis utilizing inducible deletions of PHD2 either in ECs (Phd2∆ECi) or in aSMCs (Phd2∆aSMC). Cardiovascular function and lung pathologies were studied using echocardiography, Doppler ultrasonography, intraventricular pressure measurement, histological, ultrastructural, and transcriptional methods. Cell intrinsic responses were investigated in hypoxia and in conditions mimicking hypertension-induced hemodynamic stress. Phd2∆ECi resulted in progressive pulmonary disease characterized by a thickened respiratory basement membrane (BM), alveolar fibrosis, increased pulmonary artery pressure, and adaptive hypertrophy of the right ventricle (RV). A low oxygen environment resulted in alterations in cultured ECs similar to those in Phd2∆ECi mice, involving BM components and vascular tone regulators favoring the contraction of SMCs. In contrast, Phd2∆aSMC resulted in elevated RV pressure without alterations in vascular tone regulators. Mechanistically, PHD2 inhibition in aSMCs involved  actin polymerization -related tension development via activated cofilin. The results also indicated that hemodynamic stress, rather than PHD2-dependent hypoxia response alone, potentiates structural remodeling of the extracellular matrix in the pulmonary microvasculature and respiratory failure.


Subject(s)
Hypertension, Pulmonary , Animals , Arteries/metabolism , Endothelial Cells/metabolism , Fibrosis , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice , Myocytes, Smooth Muscle/pathology , Prolyl Hydroxylases/metabolism
7.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919829

ABSTRACT

The HIF prolyl 4-hydroxylases (HIF-P4H) control hypoxia-inducible factor (HIF), a powerful mechanism regulating cellular adaptation to decreased oxygenation. The gastrointestinal epithelium subsists in "physiological hypoxia" and should therefore have an especially well-designed control over this adaptation. Thus, we assessed the absolute mRNA expression levels of the HIF pathway components, Hif1a, HIF2a, Hif-p4h-1, 2 and 3 and factor inhibiting HIF (Fih1) in murine jejunum, caecum and colon epithelium using droplet digital PCR. We found a higher expression of all these genes towards the distal end of the gastrointestinal tract. We detected mRNA for Hif-p4h-1, 2 and 3 in all parts of the gastrointestinal tract. Hif-p4h-2 had significantly higher expression levels compared to Hif-p4h-1 and 3 in colon and caecum epithelium. To test the roles each HIF-P4H isoform plays in the gut epithelium, we measured the gene expression of classical HIF target genes in Hif-p4h-1-/-, Hif-p4h-2 hypomorph and Hif-p4h-3-/- mice. Only Hif-p4h-2 hypomorphism led to an upregulation of HIF target genes, confirming a predominant role of HIF-P4H-2. However, the abundance of Hif-p4h-1 and 3 expression in the gastrointestinal epithelium implies that these isoforms may have specific functions as well. Thus, the development of selective inhibitors might be useful for diverging therapeutic needs.


Subject(s)
Gene Expression Regulation, Enzymologic , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia/enzymology , Hypoxia/genetics , Intestinal Mucosa/enzymology , Aging/metabolism , Animals , Cecum/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Isoenzymes/metabolism , Jejunum/metabolism , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
8.
PLoS One ; 15(5): e0233261, 2020.
Article in English | MEDLINE | ID: mdl-32413092

ABSTRACT

The transcription factor Hypoxia-inducible factor 1 (HIF-1) plays a pivotal role in tissue regeneration. HIF-1 is negatively controlled by O2-dependent prolyl hydroxylases with a predominant role of prolyl hydroxylase 2 isoform (Phd2). Transgenic mice, hypomorphic for this isoform, accumulate more HIF-1 under normoxic conditions. Using these mice, we investigated the influence of Phd2 and HIF-1 on the regenerative capability of skeletal muscle tissue after myotrauma. Phd2-hypomorphic and wild type mice (on C57Bl/6 background) were grouped with regeneration times from 6 to 168 hours after closed mechanic muscle trauma to the hind limb. Tissue samples were analysed by immuno-staining and real-time PCR. Bone marrow derived macrophages of wild type and Phd2-hypomorphic mice were isolated and analysed via flow cytometry and quantitative real-time PCR. Phd2 reduction led to a higher regenerative capability due to enhanced activation of myogenic factors accompanied by induction of genes responsible for glucose and lactate metabolism in Phd2-hypomorphic mice. Macrophage infiltration into the trauma areas in hypomorphic mice started earlier and was more pronounced compared to wild type mice. Phd2-hypomorphic mice also showed higher numbers of macrophages in areas with sustained trauma 72 hours after myotrauma application. In conclusion, we postulate that the HIF-1 pathway is activated secondary to a Phd2 reduction which may lead to i) higher activation of myogenic factors, ii) increased number of positive stem cell proliferation markers, and iii) accelerated macrophage recruitment to areas of trauma, resulting in faster muscle tissue regeneration after myotrauma. With the current development of prolyl hydroxylase domain inhibitors, our findings point towards a potential clinical benefit after myotrauma.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Muscle, Skeletal/physiology , Regeneration/physiology , Soft Tissue Injuries/physiopathology , Animals , Cell Proliferation/physiology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , Vascular Endothelial Growth Factor A/metabolism
9.
Matrix Biol ; 75-76: 114-125, 2019 01.
Article in English | MEDLINE | ID: mdl-29758265

ABSTRACT

The extracellular matrix (ECM) regulates numerous cellular events in addition to providing structural integrity. Among several protein and enzyme families implicated in functions of the ECM, the lysyl oxidases and ADAMTS proteins are known to participate in microfibril and elastic fiber formation as well as ECM-associated signaling. A yeast two-hybrid screen to identify lysyl oxidase (LOX) binding proteins identified ADAMTSL4 as a potential interactor. We demonstrate here that several members of the LOX and ADAMTS families interact with one another. Upon investigating the interaction between LOX and ADAMTSL2 we found that the absence or inhibition of Lox affected ADAMTSL2 molecular forms and reduced its tissue levels. Thus, ADAMTSL2 stability and inter-molecular complexes may depend on the activity of lysyl oxidases.


Subject(s)
ADAMTS Proteins/genetics , Extracellular Matrix/genetics , Multiprotein Complexes/genetics , Protein-Lysine 6-Oxidase/genetics , Animals , Elastic Tissue/chemistry , Elastic Tissue/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Humans , Mice , Microfibrils/genetics , Multiprotein Complexes/chemistry , Protein Binding , Protein Interaction Maps , Signal Transduction
10.
Sci Rep ; 7(1): 17220, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222481

ABSTRACT

Hypoxia-inducible factor 1α (HIF1α) induces the expression of several hundred genes in hypoxia aiming at restoration of oxygen homeostasis. HIF prolyl-4-hydroxylases (HIF-P4Hs) regulate the stability of HIF1α in an oxygen-dependent manner. Hypoxia is a common feature in inflammation and cancer and the HIF pathway is closely linked with the inflammatory NF-κB and tumor suppressor p53 pathways. Here we show that genetic inactivation or chemical inhibition of HIF-P4H-1 leads to downregulation of proinflammatory genes, while proapoptotic genes are upregulated. HIF-P4H-1 inactivation reduces the inflammatory response under LPS stimulus in vitro and in an acute skin inflammation model in vivo. Furthermore, HIF-P4H-1 inactivation increases p53 activity and stability and hydroxylation of proline 142 in p53 has an important role in this regulation. Altogether, our data suggest that HIF-P4H-1 inhibition may be a promising therapeutic candidate for inflammatory diseases and cancer, enhancing the reciprocal negative regulation of the NF-κB and p53 pathways.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , NF-kappa B/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cell Line , Down-Regulation , Gene Silencing , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Macrophages/cytology , Macrophages/metabolism , Mice , Proteolysis
11.
Cell Rep ; 19(4): 774-784, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445728

ABSTRACT

Surgery remains the most successful curative treatment for cancer. However, some patients with early-stage disease who undergo surgery eventually succumb to distant metastasis. Here, we show that in response to surgery, the lungs become more vulnerable to metastasis due to extracellular matrix remodeling. Mice that undergo surgery or that are preconditioned with plasma from donor mice that underwent surgery succumb to lung metastases earlier than controls. Increased lysyl oxidase (LOX) activity and expression, fibrillary collagen crosslinking, and focal adhesion signaling contribute to this effect, with the hypoxic surgical site serving as the source of LOX. Furthermore, the lungs of recipient mice injected with plasma from post-surgical colorectal cancer patients are more prone to metastatic seeding than mice injected with baseline plasma. Downregulation of LOX activity or levels reduces lung metastasis after surgery and increases survival, highlighting the potential of LOX inhibition in reducing the risk of metastasis following surgery.


Subject(s)
Colorectal Neoplasms/surgery , Lung Neoplasms/secondary , Protein-Lysine 6-Oxidase/metabolism , Animals , Antibodies/immunology , Antibodies/therapeutic use , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cell Line, Tumor , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Focal Adhesions/metabolism , Humans , Kaplan-Meier Estimate , Lung/pathology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Fluorescence , Protein-Lysine 6-Oxidase/blood , Protein-Lysine 6-Oxidase/immunology , Risk , Signal Transduction , Transplantation, Homologous
12.
Development ; 142(5): 983-93, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25715398

ABSTRACT

Muscle is an integrated tissue composed of distinct cell types and extracellular matrix. While much emphasis has been placed on the factors required for the specification of the cells that comprise muscle, little is known about the crosstalk between them that enables the development of a patterned and functional tissue. We find in mice that deletion of lysyl oxidase (Lox), an extracellular enzyme regulating collagen maturation and organization, uncouples the balance between the amount of myofibers and that of muscle connective tissue (MCT). We show that Lox secreted from the myofibers attenuates TGFß signaling, an inhibitor of myofiber differentiation and promoter of MCT development. We further demonstrate that a TGFß-Lox feedback loop between the MCT and myofibers maintains the dynamic developmental homeostasis between muscle components while also regulating MCT organization. Our results allow a better understanding of diseases such as Duchenne muscular dystrophy, in which LOX and TGFß signaling have been implicated and the balance between muscle constituents is disturbed.


Subject(s)
Extracellular Matrix Proteins/metabolism , Muscles/embryology , Muscles/metabolism , Protein-Lysine 6-Oxidase/metabolism , Transforming Growth Factor beta/metabolism , Animals , Connective Tissue/embryology , Connective Tissue/metabolism , Connective Tissue/ultrastructure , Extracellular Matrix Proteins/genetics , Female , Immunohistochemistry , In Situ Hybridization , Mice , Microscopy, Electron, Transmission , Muscles/ultrastructure , Pregnancy , Protein-Lysine 6-Oxidase/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/genetics
13.
Blood ; 120(16): 3336-44, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22955912

ABSTRACT

An endoplasmic reticulum transmembrane prolyl 4-hydroxylase (P4H-TM) is able to hydroxylate the α subunit of the hypoxia-inducible factor (HIF) in vitro and in cultured cells, but nothing is known about its roles in mammalian erythropoiesis. We studied such roles here by administering a HIF-P4H inhibitor, FG-4497, to P4h-tm(-/-) mice. This caused larger increases in serum Epo concentration and kidney but not liver Hif-1α and Hif-2α protein and Epo mRNA levels than in wild-type mice, while the liver Hepcidin mRNA level was lower in the P4h-tm(-/-) mice than in the wild-type. Similar, but not identical, differences were also seen between FG-4497-treated Hif-p4h-2 hypomorphic (Hif-p4h-2(gt/gt)) and Hif-p4h-3(-/-) mice versus wild-type mice. FG-4497 administration increased hemoglobin and hematocrit values similarly in the P4h-tm(-/-) and wild-type mice, but caused higher increases in both values in the Hif-p4h-2(gt/gt) mice and in hematocrit value in the Hif-p4h-3(-/-) mice than in the wild-type. Hif-p4h-2(gt/gt)/P4h-tm(-/-) double gene-modified mice nevertheless had increased hemoglobin and hematocrit values without any FG-4497 administration, although no such abnormalities were seen in the Hif-p4h-2(gt/gt) or P4h-tm(-/-) mice. Our data thus indicate that P4H-TM plays a role in the regulation of EPO production, hepcidin expression, and erythropoiesis.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/blood , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Procollagen-Proline Dioxygenase/physiology , Animals , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Hematocrit , Hemoglobins/metabolism , Hepcidins , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Procollagen-Proline Dioxygenase/antagonists & inhibitors , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
14.
Mol Cell Biol ; 31(13): 2683-95, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21536655

ABSTRACT

The lysyl oxidase gene (LOX) inhibits Ras signaling in transformed fibroblasts and breast cancer cells. Its activity was mapped to the 162-amino-acid propeptide domain (LOX-PP) of the lysyl oxidase precursor protein. LOX-PP inhibits Erk signaling, motility, and tumor formation in a breast cancer xenograft model; however, its mechanism of action is largely unknown. Here, a copurification-mass spectrometry approach was taken using ectopically expressed LOX-PP in HEK293T cells and the heat shock/chaperone protein Hsp70 identified. Hsp70 interaction with LOX-PP was confirmed using coimmunoprecipitation of intracellularly and bacterially expressed and endogenous proteins. The interaction was mapped to the Hsp70 peptide-binding domain and to LOX-PP amino acids 26 to 100. LOX-PP association reduced Hsp70 chaperone activities of protein refolding and survival after heat shock. LOX-PP interacted with the Hsp70 chaperoned protein c-Raf. With the use of ectopic expression of LOX-PP wild-type and deletion proteins, small interfering RNA (siRNA) knockdown, and Lox(-/-) mouse embryo fibroblasts, LOX-PP interaction with c-Raf was shown to decrease downstream activation of MEK and NF-κB, migration, and anchorage-independent growth and reduce its mitochondrial localization. Thus, the interaction of LOX-PP with Hsp70 and c-Raf inhibits a critical intermediate in Ras-induced MEK signaling and plays an important role in the function of this tumor suppressor.


Subject(s)
Breast Neoplasms/metabolism , HSP70 Heat-Shock Proteins/metabolism , Protein-Lysine 6-Oxidase/metabolism , ras Proteins/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , Fibroblasts/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Mass Spectrometry , Mice , Mitochondria/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Structure, Tertiary , Protein-Lysine 6-Oxidase/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering/genetics
15.
J Neural Transm (Vienna) ; 118(7): 1091-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21509606

ABSTRACT

Lysyl oxidase (LOX) is a potent chemokine inducing the migration of varied cell types. Here we demonstrate that inhibition of cellular LOX activity by preincubation of vascular smooth muscle cells (VSMC) with ß-aminopropionitrile (BAPN), the irreversible inhibitor of LOX activity, resulted in the marked suppression of the chemotactic response and sensitivity of these cells toward LOX and toward PDGF-BB. Plasma membranes purified from VSMC not previously exposed to BAPN contained a group of oxidized plasma membrane proteins, including the PDGF receptor, PDGFR-ß. The oxidation of this receptor and other membrane proteins was largely prevented in cells preincubated with BAPN. Addition of purified LOX to BAPN-free cells, which had been previously exposed to BAPN, restored the profile of oxidized proteins towards that of control cells. The high affinity and capacity for the binding of PDGF-BB by cells was significantly diminished when compared with cells in which oxidation by LOX was prevented by BAPN. The chemotactic responses of LOX knock-out mouse embryonic fibroblasts mirrored those obtained with VSMC treated with BAPN. These novel findings suggest that LOX activity is essential to generate optimal chemotactic sensitivity of cells to chemoattractants by oxidizing specific cell surface proteins, such as PDGFR-ß.


Subject(s)
Cell Membrane/enzymology , Chemokines/metabolism , Chemotaxis/physiology , Membrane Proteins/metabolism , Protein-Lysine 6-Oxidase/metabolism , Animals , Cattle , Cell Membrane/metabolism , Cells, Cultured , Chemokines/agonists , Chemokines/physiology , Chemotaxis/drug effects , Mice , Mice, Knockout , Oxidation-Reduction , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Protein-Lysine 6-Oxidase/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/physiology
16.
J Biol Chem ; 285(18): 13646-57, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20185832

ABSTRACT

Hypoxia-inducible factor (HIF) has a pivotal role in oxygen homeostasis and cardioprotection mediated by ischemic preconditioning. Its stability is regulated by HIF prolyl 4-hydroxylases (HIF-P4Hs), the inhibition of which is regarded as a promising strategy for treating diseases such as anemia and ischemia. We generated a viable Hif-p4h-2 hypomorph mouse line (Hif-p4h-2(gt/gt)) that expresses decreased amounts of wild-type Hif-p4h-2 mRNA: 8% in the heart; 15% in the skeletal muscle; 34-47% in the kidney, spleen, lung, and bladder; 60% in the brain; and 85% in the liver. These mice have no polycythemia and show no signs of the dilated cardiomyopathy or hyperactive angiogenesis observed in mice with broad spectrum conditional Hif-p4h-2 inactivation. We focused here on the effects of chronic Hif-p4h-2 deficiency in the heart. Hif-1 and Hif-2 were stabilized, and the mRNA levels of glucose transporter-1, several enzymes of glycolysis, pyruvate dehydrogenase kinase 1, angiopoietin-2, and adrenomedullin were increased in the Hif-p4h-2(gt/gt) hearts. When isolated Hif-p4h-2(gt/gt) hearts were subjected to ischemia-reperfusion, the recovery of mechanical function and coronary flow rate was significantly better than in wild type, while cumulative release of lactate dehydrogenase reflecting the infarct size was reduced. The preischemic amount of lactate was increased, and the ischemic versus preischemic [CrP]/[Cr] and [ATP] remained at higher levels in Hif-p4h-2(gt/gt) hearts, indicating enhanced glycolysis and an improved cellular energy state. Our data suggest that chronic stabilization of Hif-1alpha and Hif-2alpha by genetic knockdown of Hif-p4h-2 promotes cardioprotection by induction of many genes involved in glucose metabolism, cardiac function, and blood pressure.


Subject(s)
Dioxygenases/metabolism , Glucose/metabolism , Muscle Proteins/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Acute Disease , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Coronary Circulation/genetics , Dioxygenases/genetics , Glucose/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Mice , Mice, Transgenic , Muscle Proteins/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Organ Specificity/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
17.
Histol Histopathol ; 24(5): 651-60, 2009 05.
Article in English | MEDLINE | ID: mdl-19283672

ABSTRACT

Lysyl oxidase (LOX) catalyzes the oxidation of the side chain of a peptidyl lysine converting specific lysine and hydroxylysine residues of alpha-aminoadipic-delta-semialdehydes, which form covalent crosslinks in collagens and elastin. Five different but closely related lysyl oxidase isoenzymes have been identified to date, and they seem to have overlapping functions in many tissues. Modification of the extracellular matrix by lysyl oxidases has been shown to be a critical contributor to the development of various organs and certain pathological conditions.


Subject(s)
Protein-Lysine 6-Oxidase/metabolism , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Animals , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/etiology , Cardiovascular System/enzymology , Cardiovascular System/growth & development , Humans , Mammals , Models, Biological , Neoplasms/enzymology , Neoplasms/etiology , Protein-Lysine 6-Oxidase/genetics , Respiratory System/enzymology , Respiratory System/growth & development
18.
J Biol Chem ; 283(35): 24103-17, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18586678

ABSTRACT

Lysyl oxidase (LOX) is a potent chemokine inducing the migration of varied cell types. Here we demonstrate that inhibition of LOX activity by beta-aminopropionitrile (BAPN) in cultured rat aortic smooth muscle cells (SMCs) reduced the chemotactic response and sensitivity of these cells toward LOX and toward PDGF-BB. The chemotactic activity of PDGF-BB was significantly enhanced in the presence of a non-chemotactic concentration of LOX. We considered the possibility that extracellular LOX may oxidize cell surface proteins, including the PDGF receptor-beta (PDGFR-beta), to affect PDGF-BB-induced chemotaxis. Plasma membranes purified from control SMC contained oxidized PDGFR-beta. The oxidation of this receptor and other membrane proteins was largely prevented in cells preincubated with BAPN. Addition of purified LOX to these cells restored the profile of oxidized proteins toward that of control cells. The high affinity and capacity for the binding of PDGF-BB by cells containing oxidized PDGFR-beta was diminished by approximately 2-fold when compared with cells in which oxidation by LOX was prevented by BAPN. Phosphorylated members of the PDGFR-beta-dependent signal transduction pathway, including PDGFR-beta, SHP2, AKT1, and ERK1/ERK2 (p44/42 MAPK), turned over faster in BAPN-treated than in control SMCs. LOX knock-out mouse embryonic fibroblasts mirrored the effect obtained with SMCs treated with BAPN. These novel findings suggest that LOX activity is essential to generate optimal chemotactic sensitivity of cells to chemoattractants by oxidizing specific cell surface proteins, such as PDGFR-beta.


Subject(s)
Aorta/enzymology , Chemotaxis/physiology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Protein-Lysine 6-Oxidase/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Aminopropionitrile/pharmacology , Animals , Aorta/cytology , Becaplermin , Cells, Cultured , Chemotaxis/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Fibroblasts/cytology , Fibroblasts/enzymology , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Oxidation-Reduction , Platelet-Derived Growth Factor/pharmacology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Proto-Oncogene Proteins c-sis , Rats , Receptor, Platelet-Derived Growth Factor beta/genetics
19.
Cancer Res ; 67(9): 4123-9, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17456585

ABSTRACT

Promoter hypermethylation is one of the common mechanisms leading to gene silencing in various human cancers. Using a combination of pharmacologic unmasking and microarray techniques, we identified 59 candidate hypermethylated genes, including LOXL1, a lysyl oxidase-like gene, in human bladder cancer cells. We further showed that LOXL1 and LOXL4 are commonly silenced genes in human bladder cancer cells, and this silence is predominantly related to promoter methylation. We also found LOXL1 and LOXL4 gene methylation and loss of expression in primary bladder tumors. In addition, somatic mutations were identified in LOXL4, but not in LOXL1 in bladder cancer. Moreover, reintroduction of LOXL1 and LOXL4 genes into human bladder cancer cells leads to a decrease of colony formation ability. Further studies indicated that the overexpression of LOXL1 and LOXL4 could antagonize Ras in activating the extracellular signal-regulated kinase (ERK) signaling pathway. Thus, our current study suggests for the first time that lysyl oxidase-like genes can act as tumor suppressor genes and exert their functions through the inhibition of the Ras/ERK signaling pathway in human bladder cancer.


Subject(s)
Amino Acid Oxidoreductases/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Urinary Bladder Neoplasms/genetics , ras Proteins/antagonists & inhibitors , Amino Acid Oxidoreductases/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Cytoplasm/enzymology , DNA Methylation/drug effects , Decitabine , Epigenesis, Genetic , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , MAP Kinase Signaling System , Mutation , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Protein-Lysine 6-Oxidase , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , ras Proteins/metabolism
20.
Am J Pathol ; 167(4): 927-36, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16192629

ABSTRACT

Lysyl oxidases, a family comprising LOX and four LOX-like enzymes, catalyze crosslinking of elastin and collagens. Mouse Lox was recently shown to be crucial for development of the cardiovascular system because null mice died perinatally of aortic aneurysms and cardiovascular dysfunction. We show here that Lox is also essential for development of the respiratory system and the integrity of elastic and collagen fibers in the lungs and skin. The lungs of E18.5 Lox(-/-) embryos showed impaired development of the distal and proximal airways. Elastic fibers in E18.5 Lox(-/-) lungs were markedly less intensely stained and more disperse than in the wild type, especially in the mesenchyme surrounding the distal airways, bronchioles, bronchi, and trachea, and were fragmented in pulmonary arterial walls. The organization of individual collagen fibers into tight bundles was likewise abnormal. Similar elastic and collagen fiber abnormalities were seen in the skin. Lysyl oxidase activity in cultured Lox(-/-) skin fibroblasts and aortic smooth muscle cells was reduced by approximately 80%, indicating that Lox is the main isoenzyme in these cells. LOX abnormalities may thus be critical for the pathogenesis of several common diseases, including pulmonary, skin, and cardiovascular disorders.


Subject(s)
Collagen/metabolism , Elastin/metabolism , Protein-Lysine 6-Oxidase/physiology , Respiratory System/growth & development , Respiratory System/metabolism , Animals , Aorta/cytology , Aorta/embryology , Cells, Cultured , Collagen/ultrastructure , Collagen Type I/metabolism , Collagen Type I/ultrastructure , Collagen Type IV/metabolism , Collagen Type IV/ultrastructure , Culture Media, Conditioned/analysis , Elastin/ultrastructure , Embryonic Development , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Heterozygote , Homozygote , Immunohistochemistry , Lung/embryology , Lung/enzymology , Lung/growth & development , Lung/metabolism , Lung/ultrastructure , Mice , Mice, Knockout , Microscopy, Fluorescence , Microscopy, Immunoelectron , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Protein-Lysine 6-Oxidase/analysis , Protein-Lysine 6-Oxidase/genetics , Respiratory System/embryology , Respiratory System/enzymology , Respiratory System/ultrastructure , Rhodamines , Skin/cytology , Skin/embryology , Skin/enzymology , Skin/growth & development , Skin/metabolism , Skin/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...