Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
2.
Semin Cell Dev Biol ; 53: 155-67, 2016 05.
Article in English | MEDLINE | ID: mdl-26768548

ABSTRACT

The fibroblast growth factors (FGFs) are a family of cell intrinsic regulatory peptides that control a broad spectrum of cellular activities. The family includes canonic FGFs that elicit their activities by activating the FGF receptor (FGFR) tyrosine kinase and non-canonic members that elicit their activities intracellularly and via FGFR-independent mechanisms. The FGF signaling axis is highly complex due to the existence of multiple isoforms of both ligands and receptors, as well as cofactors that include the chemically heterogeneous heparan sulfate (HS) cofactors, and in the case of endocrine FGFs, the Klotho coreceptors. Resident FGF signaling controls embryonic development, maintains tissue homeostasis, promotes wound healing and tissue regeneration, and regulates functions of multiple organs. However, ectopic or aberrant FGF signaling is a culprit for various diseases, including congenital birth defects, metabolic disorder, and cancer. The molecular mechanisms by which the specificity of FGF signaling is achieved remain incompletely understood. Since its application as a druggable target has been gradually recognized by pharmaceutical companies and translational researchers, understanding the determinants of FGF signaling specificity has become even more important in order to get into the position to selectively suppress a particular pathway without affecting others to minimize side effects.


Subject(s)
Fibroblast Growth Factors/metabolism , Animals , Humans , Models, Biological , Neoplasms/metabolism , Signal Transduction , Translational Research, Biomedical
3.
J Biol Chem ; 291(3): 1243-50, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26565030

ABSTRACT

Phagocytosis is a critical cellular process for innate immune defense against microbial infection. The regulation of phagocytosis process is complex and has not been well defined. An intracellular molecule might regulate cell surface-initiated phagocytosis, but the underlying molecular mechanism is poorly understood (1). In this study, we found that microtubule-associated protein 1S (MAP1S), a protein identified recently that is involved in autophagy (2), is expressed primarily in macrophages. MAP1S-deficient macrophages are impaired in the phagocytosis of bacteria. Furthermore, we demonstrate that MAP1S interacts directly with MyD88, a key adaptor of Toll-like receptors (TLRs), upon TLR activation and affects the TLR signaling pathway. Intriguingly, we also observe that, upon TLR activation, MyD88 participates in autophagy processing in a MAP1S-dependent manner by co-localizing with MAP1 light chain 3 (MAP1-LC3 or LC3). Therefore, we reveal that an intracellular autophagy-related molecule of MAP1S controls bacterial phagocytosis through TLR signaling.


Subject(s)
Macrophages/metabolism , Microtubule-Associated Proteins/metabolism , Myeloid Differentiation Factor 88/agonists , Phagocytosis , Salmonella typhimurium/immunology , Signal Transduction , Staphylococcus aureus/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Cells, Cultured , HEK293 Cells , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Transport , RAW 264.7 Cells , Specific Pathogen-Free Organisms , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
4.
J Biol Chem ; 290(29): 17753-17761, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26032417

ABSTRACT

Prostate stem cells (P-SCs) are capable of giving rise to all three lineages of prostate epithelial cells, which include basal, luminal, and neuroendocrine cells. Two types of P-SCs have been identified in both human and mouse adult prostates based on prostasphere or organoid cultures, cell lineage tracing, renal capsule implantation, and expression of luminal- and basal-specific proteins. The sphere-forming P-SCs are from the basal cell compartment that express P63, and are therefore designated as basal P-SCs (P-bSCs). Luminal P-SCs (P-lSCs) express luminal cytokeratins and Nkx3.1. Herein, we report that the type 2 FGF receptor (FGFR2) signaling axis is crucial for preserving stemness and preventing differentiation of P-bSCs. FGFR2 signaling mediated by FGFR substrate 2α (FRS2α) is indispensable for formation and maintenance of prostaspheres derived from P63(+) P-bSCs. Ablation of Fgfr2 in P63(+) cells in vitro causes the disintegration of prostaspheres. Ablation of Fgfr2 in vivo reduces the number of P63-expressing basal cells and enriches luminal cells. This suggests a basal stem cell-to-luminal cell differentiation. In addition, ablation of Fgfr2 in P63(+) cells causes defective postnatal development of the prostate. Therefore, the data indicate that FGFR2 signaling is critical for preserving stemness and preventing differentiation of P-bSCs.


Subject(s)
Adult Stem Cells/cytology , Prostate/cytology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Male , Mice , Phosphoproteins/analysis , Prostate/metabolism , Prostate/ultrastructure , Spheroids, Cellular , Trans-Activators/analysis
5.
J Biol Chem ; 290(29): 17745-17752, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26032419

ABSTRACT

Prostate stem cells (P-SCs) are capable of giving rise to all three lineages of prostate epithelial cells, including basal, luminal, and neuroendocrine cells. Multiple methods have been used to identify P-SCs in adult prostates. These include in vivo renal capsule implantation of a single epithelial cell with urogenital mesenchymal cells, in vitro prostasphere and organoid cultures, and lineage tracing with castration-resistant Nkx3.1 expression (CARN), in conjunction with expression of cell type-specific markers. Both organoid culture and CARN tracing show the existence of P-SCs in the luminal compartment. Although prostasphere cells predominantly express basal cell-specific cytokeratin and P63, the lineage of prostasphere-forming cells in the P-SC hierarchy remains to be determined. Using lineage tracing with P63(CreERT2), we show here that the sphere-forming P-SCs are P63-expressing cells and reside in the basal compartment. Therefore we designate them as basal P-SCs (P-bSCs). P-bSCs are capable of differentiating into AR(+) and CK18(+) organoid cells, but organoid cells cannot form spheres. We also report that prostaspheres contain quiescent stem cells. Therefore, the results show that P-bSCs represent stem cells that are early in the hierarchy of overall prostate tissue stem cells. Understanding the contribution of the two types of P-SCs to prostate development and prostate cancer stem cells and how to manipulate them may open new avenues for control of prostate cancer progression and relapse.


Subject(s)
Adult Stem Cells/cytology , Phosphoproteins/analysis , Prostate/cytology , Trans-Activators/analysis , Animals , Cell Differentiation , Cells, Cultured , Epithelial Cells/cytology , Homeodomain Proteins/analysis , Male , Mice , Organ Culture Techniques , Receptors, G-Protein-Coupled/analysis , Spheroids, Cellular , Transcription Factors/analysis
6.
PLoS Biol ; 10(5): e1001326, 2012.
Article in English | MEDLINE | ID: mdl-22589701

ABSTRACT

Elevated aerobic glycolysis in cancer cells (the Warburg effect) may be attributed to respiration injury or mitochondrial dysfunction, but the underlying mechanisms and therapeutic significance remain elusive. Here we report that induction of mitochondrial respiratory defect by tetracycline-controlled expression of a dominant negative form of DNA polymerase ƎĀ³ causes a metabolic shift from oxidative phosphorylation to glycolysis and increases ROS generation. We show that upregulation of NOX is critical to support the elevated glycolysis by providing additional NAD+. The upregulation of NOX is also consistently observed in cancer cells with compromised mitochondria due to the activation of oncogenic Ras or loss of p53, and in primary pancreatic cancer tissues. Suppression of NOX by chemical inhibition or genetic knockdown of gene expression selectively impacts cancer cells with mitochondrial dysfunction, leading to a decrease in cellular glycolysis, a loss of cell viability, and inhibition of cancer growth in vivo. Our study reveals a previously unrecognized function of NOX in cancer metabolism and suggests that NOX is a potential novel target for cancer treatment.


Subject(s)
Glycolysis , Mitochondria/pathology , NADPH Oxidases/metabolism , Pancreatic Neoplasms/enzymology , Animals , Cell Survival , Enzyme Activation , Gene Knockdown Techniques , Genes, Neoplasm , HEK293 Cells , Humans , Mice , Mice, Nude , Mitochondria/enzymology , Mitochondria/metabolism , NADPH Oxidase 1 , NADPH Oxidases/genetics , Oxidative Phosphorylation , Pancreatic Neoplasms/pathology , Plasmids/genetics , Plasmids/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tetracycline/pharmacology , Transfection , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 288(30): 22174-83, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23754280

ABSTRACT

Cleft palate is a common congenital birth defect. The fibroblast growth factor (FGF) family has been shown to be important for palatogenesis, which elicits the regulatory functions by activating the FGF receptor tyrosine kinase. Mutations in Fgf or Fgfr are associated with cleft palate. To date, most mechanistic studies on FGF signaling in palate development have focused on FGFR2 in the epithelium. Although Fgfr1 is expressed in the cranial neural crest (CNC)-derived palate mesenchyme and Fgfr1 mutations are associated with palate defects, how FGFR1 in palate mesenchyme regulates palatogenesis is not well understood. Here, we reported that by using Wnt1(Cre) to delete Fgfr1 in neural crest cells led to cleft palate, cleft lip, and other severe craniofacial defects. Detailed analyses revealed that loss-of-function mutations in Fgfr1 did not abrogate patterning of CNC cells in palate shelves. However, it upset cell signaling in the frontofacial areas, delayed cell proliferation in both epithelial and mesenchymal compartments, prevented palate shelf elevation, and compromised palate shelf fusion. This is the first report revealing how FGF signaling in CNC cells regulates palatogenesis.


Subject(s)
Cleft Palate/metabolism , Mesoderm/metabolism , Neural Crest/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cell Proliferation , Cleft Palate/embryology , Cleft Palate/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Epithelium/embryology , Epithelium/metabolism , Gene Expression Regulation, Developmental , Immunohistochemistry , Lac Operon/genetics , Mesoderm/embryology , Mice , Mice, Knockout , Mice, Transgenic , Neural Crest/cytology , Neural Crest/embryology , Palate/embryology , Palate/metabolism , Palate/pathology , Proteins/genetics , Proteins/metabolism , RNA, Untranslated , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Time Factors , Tissue Culture Techniques
8.
J Biol Chem ; 288(40): 28952-61, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23979135

ABSTRACT

A constant supply of epithelial cells from dental epithelial stem cell (DESC) niches in the cervical loop (CL) enables mouse incisors to grow continuously throughout life. Elucidation of the cellular and molecular mechanisms underlying this unlimited growth potential is of broad interest for tooth regenerative therapies. Fibroblast growth factor (FGF) signaling is essential for the development of mouse incisors and for maintenance of the CL during prenatal development. However, how FGF signaling in DESCs controls the self-renewal and differentiation of the cells is not well understood. Herein, we report that FGF signaling is essential for self-renewal and the prevention of cell differentiation of DESCs in the CL as well as in DESC spheres. Inhibiting the FGF signaling pathway decreased proliferation and increased apoptosis of the cells in DESC spheres. Suppressing FGFR or its downstream signal transduction pathways diminished Lgr5-expressing cells in the CL and promoted cell differentiation both in DESC spheres and the CL. Furthermore, disruption of the FGF pathway abrogated Wnt signaling to promote Lgr5 expression in DESCs both in vitro and in vivo. This study sheds new light on understanding the mechanism by which the homeostasis, expansion, and differentiation of DESCs are regulated.


Subject(s)
Epithelial Cells/cytology , Fibroblast Growth Factors/metabolism , Signal Transduction , Stem Cells/cytology , Tooth/cytology , Animals , Cell Cycle , Cell Differentiation , Cell Proliferation , Epithelial Cells/enzymology , MAP Kinase Signaling System , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, G-Protein-Coupled/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Stem Cells/enzymology , Up-Regulation , Wnt Proteins/metabolism
9.
Pflugers Arch ; 466(5): 1011-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24068255

ABSTRACT

Cholangiocytes, bile duct lining cells, actively adjust the amount of cholesterol and bile acids in bile through expression of enzymes and channels involved in transportation and metabolism of the cholesterol and bile acids. Herein, we report molecular mechanisms regulating bile acid biosynthesis in cholangiocytes. Among the cytochrome p450 (Cyp) enzymes involved in bile acid biosynthesis, sterol 27-hydroxylase (Cyp27) that is the rate-limiting enzyme for the acidic pathway of bile acid biosynthesis expressed in cholangiocytes. Expression of other Cyp enzymes for the basic bile acid biosynthesis was hardly detected. The Cyp27 expression was negatively regulated by a hydrophobic bile acid through farnesoid X receptor (FXR), a nuclear receptor activated by bile acid ligands. Activated FXR exerted the negative effects by inducing an expression of fibroblast growth factor 15/19 (FGF15/19). Similar to its repressive function against cholesterol 7α-hydroxylase (Cyp7a1) expression in hepatocytes, secreted FGF15/19 triggered Cyp27 repression in cholangiocytes through interaction with its cognate receptor fibroblast growth factor receptor 4 (FGFR4). The involvements of FXR and FGFR4 for the bile acid-induced Cyp27 repression were confirmed in vivo using knockout mouse models. Different from the signaling in hepatocytes, wherein the FGF15/19-induced repression signaling is mediated by c-Jun N-terminal kinase (JNK), FGF15/19-induced Cyp27 repression in cholangiocytes was mediated by p38 kinase. Thus, the results collectively suggest that cholangiocytes may be able to actively regulate bile acid biosynthesis in cholangiocytes and even hepatocyte by secreting FGF15/19. We suggest the presence of cholangiocyte-mediated intrahepatic feedback loop in addition to the enterohepatic feedback loop against bile acid biosynthesis in the liver.


Subject(s)
Bile Ducts/metabolism , Cytochrome P-450 Enzyme System/metabolism , Epithelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bile Acids and Salts/metabolism , Bile Ducts/cytology , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Fibroblast Growth Factors/genetics , Hep G2 Cells , Humans , Mice , Rats , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
10.
Circ Res ; 110(4): e29-39, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22207710

ABSTRACT

RATIONALE: Although the fibroblast growth factor (FGF) signaling axis plays important roles in heart development, the molecular mechanism by which the FGF regulates cardiogenesis is not fully understood. OBJECTIVE: To investigate the mechanism by which FGF signaling regulates cardiac progenitor cell differentiation. METHODS AND RESULTS: Using mice with tissue-specific ablation of FGF receptors and FGF receptor substrate 2α (Frs2α) in heart progenitor cells, we demonstrate that disruption of FGF signaling leads to premature differentiation of cardiac progenitor cells in mice. Using embryoid body cultures of mouse embryonic stem cells, we reveal that FGF signaling promotes mesoderm differentiation in embryonic stem cells but inhibits cardiomyocyte differentiation of the mesoderm cells at later stages. Furthermore, we also report that inhibiting FRS2α-mediated signals increases autophagy and that activating autophagy promotes myocardial differentiation and vice versa. CONCLUSIONS: The results indicate that the FGF/FRS2α-mediated signals prevent premature differentiation of heart progenitor cells through suppressing autophagy. The findings provide the first evidence that autophagy plays a role in heart progenitor differentiation.


Subject(s)
Autophagy , Cell Differentiation , Fibroblast Growth Factors/metabolism , Heart/embryology , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Embryo Culture Techniques , Gene Expression Regulation, Developmental , Genotype , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Phenotype , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stem Cells/pathology , Time Factors , Tissue Culture Techniques
11.
FASEB J ; 26(5): 2105-16, 2012 May.
Article in English | MEDLINE | ID: mdl-22278938

ABSTRACT

We have previously found that in failing human hearts, Rho-associated coiled-coil protein kinase 1 (ROCK1) is processed by caspase-3 into an active isoform, ROCKΔ1. The purpose of the current investigation was to elucidate the pathological consequences of truncated ROCK1 accumulation in the heart, the associated molecular mechanism of ROCKΔ1-mediated cardiac phenotype, and the molecular signaling between Rho kinase activation in cardiomyocytes and extracellular matrix response. We generated transgenic mice expressing ROCKΔ1 in cardiomyocytes to mimic the situation observed in human heart disease, whereas an additional kinase-deficient mouse was generated as a control. The ROCKΔ1 transgenic mice developed fibrotic cardiomyopathy with diastolic dysfunction. Transgenic hearts displayed activated TGFƟ1 and NF-κB signaling and a release of a subset of cytokines and were susceptible to angiotensin II stress. Treatment with a Rho kinase inhibitor attenuated the fibrotic phenotype. Cardiac fibroblasts differentiated into myofibroblasts when cocultured with transgenic cardiomyocytes but not with wild-type cardiomyocytes. Inhibitors of Rho kinase as well as TGFƟR1 and NF-κB decreased these effects. The serum response factor-dependent TGFƟ1 regulation was shown to be responsible for the Rho kinase-mediated activation of TGFƟ1 signaling. We conclude that ROCKΔ1 is a novel fibrotic factor. Activation of TGFƟ1 and NF-κB signaling contributes to the Rho kinase-mediated pathological fibrosis.


Subject(s)
Cardiomyopathies/enzymology , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers , Fibrosis , Mice , Mice, Transgenic , Polymerase Chain Reaction , Rats , Transforming Growth Factor beta1/metabolism , rho-Associated Kinases/metabolism
12.
BMC Gastroenterol ; 13: 67, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23590285

ABSTRACT

BACKGROUND: FGF21 is a promising intervention therapy for metabolic diseases as fatty liver, obesity and diabetes. Recent results suggest that FGF21 is highly expressed in hepatocytes under metabolic stress caused by starvation, hepatosteatosis, obesity and diabetes. Hepatic FGF21 elicits metabolic benefits by targeting adipocytes of the peripheral adipose tissue through the transmembrane FGFR1-KLB complex. Ablation of adipose FGFR1 resulted in increased hepatosteatosis under starvation conditions and abrogation of the anti-obesogenic action of FGF21. These results indicate that FGF21 may be a stress responsive hepatokine that targets adipocytes and adipose tissue for alleviating the damaging effects of stress on the liver. However, it is unclear whether hepatic induction of FGF21 is limited to only metabolic stress, or to a more general hepatic stress resulting from liver pathogenesis and injury. METHODS: In this survey-based study, we examine the nature of hepatic FGF21 activation in liver tissues and tissue sections from several mouse liver disease models and human patients, by quantitative PCR, immunohistochemistry, protein chemistry, and reporter and CHIP assays. The liver diseases include genetic and chemical-induced HCC, liver injury and regeneration, cirrhosis, and other types of liver diseases. RESULTS: We found that mouse FGF21 is induced in response to chemical (DEN treatment) and genetic-induced hepatocarcinogenesis (disruptions in LKB1, p53, MST1/2, SAV1 and PTEN). It is also induced in response to loss of liver mass due to partial hepatectomy followed by regeneration. The induction of FGF21 expression is potentially under the control of stress responsive transcription factors p53 and STAT3. Serum FGF21 levels correlate with FGF21 expression in hepatocytes. In patients with hepatitis, fatty degeneration, cirrhosis and liver tumors, FGF21 levels in hepatocytes or phenotypically normal hepatocytes are invariably elevated compared to normal health subjects. CONCLUSION: FGF21 is an inducible hepatokine and could be a biomarker for normal hepatocyte function. Activation of its expression is a response of functional hepatocytes to a broad spectrum of pathological changes that impose both cellular and metabolic stress on the liver. Taken together with our recent data, we suggest that hepatic FGF21 is a general stress responsive factor that targets adipose tissue for normalizing local and systemic metabolic parameters while alleviating the overload and damaging effects imposed by the pathogenic stress on the liver. This study therefore provides a rationale for clinical biomarker studies in humans.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Fibroblast Growth Factors/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , AMP-Activated Protein Kinases , Animals , Carcinoma, Hepatocellular/chemically induced , Cell Transformation, Neoplastic/genetics , Diethylnitrosamine , Disease Models, Animal , Fibroblast Growth Factors/genetics , Hepatocytes/metabolism , Humans , Klotho Proteins , Liver/metabolism , Liver/pathology , Liver/surgery , Liver Neoplasms/chemically induced , Male , Membrane Proteins/genetics , Mice , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , STAT3 Transcription Factor/metabolism , Stress, Physiological , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
J Biol Chem ; 286(12): 10367-77, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21262964

ABSTRACT

The ubiquitously distributed MAP1S is a homologue of the exclusively neuronal distributed microtubule-associated protein 1A and 1B (MAP1A/B). They give rise to multiple isoforms through similar post-translational modification. Isoforms of MAP1S have been implicated in microtubule dynamics and mitotic abnormalities and mitotic cell death. Here we show that ablation of the Map1s gene in mice caused reduction in the B-cell CLL/lymphoma 2 or xL (Bcl-2/xL) and cyclin-dependent kinase inhibitor 1B (P27) protein levels, accumulation of defective mitochondria, and severe defects in response to nutritive stress, suggesting defects in autophagosomal biogenesis and clearance. Furthermore, MAP1S isoforms interacted with the autophagosome-associated light chain 3 of MAP1A/B (LC3), a homologue of yeast autophagy-related gene 8 (ATG8), and recruited it to stable microtubules in a MAP1S and LC3 isoform-dependent mode. In addition, MAP1S interacted with mitochondrion-associated leucine-rich PPR-motif containing protein (LRPPRC) that interacts with the mitophagy initiator and Parkinson disease-related protein Parkin. The three-way interactions of MAP1S isoforms with LC3 and microtubules as well as the interaction of MAP1S with LRPPRC suggest that MAP1S isoforms may play positive roles in integration of autophagic components with microtubules and mitochondria in both autophagosomal biogenesis and degradation. For the first time, our results clarify roles of MAP1S in bridging microtubules and mitochondria with autophagic and mitophagic initiation, maturation, trafficking, and lysosomal clearance. Defects in the MAP1S-regulated autophagy may impact heart disease, cancers, neurodegenerative diseases, and a wide range of other diseases.


Subject(s)
Autophagy/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitochondria/metabolism , Phagosomes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs/physiology , Animals , Autophagy-Related Protein 8 Family , HEK293 Cells , Heart Diseases/genetics , Heart Diseases/metabolism , Humans , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Mitochondria/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Phagosomes/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
14.
J Biol Chem ; 285(39): 30069-78, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20657013

ABSTRACT

In organs involved in metabolic homeostasis, transmembrane α and Ɵklothos direct FGFR signaling to control of metabolic pathways. Coordinate expression of Ɵklotho and FGFR4 is a property of mature hepatocytes. Genetic deletion of FGFR4 or Ɵklotho in mice disrupts hepatic cholesterol/bile acid and lipid metabolism. The deletion of FGFR4 has no effect on the proliferative response of hepatocytes after liver injury. However, its absence results in accelerated progression of dimethynitrosamine-initiated hepatocellular carcinomas, indicating that FGFR4 suppresses hepatoma proliferation. The mechanism underlying the FGFR4-mediated hepatoma suppression has not been addressed. Here we show that Ɵklotho expression is more consistently down-regulated in human and mouse hepatomas than FGFR4. Co-expression and activation by either endocrine FGF19 or cellular FGF1 of the FGFR4 kinase in a complex with Ɵklotho restricts cell population growth through induction of apoptotic cell death in both hepatic and nonhepatic cells. The Ɵklotho-FGFR4 partnership caused a depression of activated AKT and mammalian target of rapamycin while activating ERK1/2 that may underlie the pro-apoptotic effect. Our results show that Ɵklotho not only interacts with heparan sulfate-FGFR4 to form a complex with high affinity for endocrine FGF19 but also impacts the quality of downstream signaling and biological end points activated by either FGF19 or canonical FGF1. Thus the same Ɵklotho-heparan sulfate-FGFR4 partnership that mediates endocrine control of hepatic metabolism plays a role in cellular homeostasis and hepatoma suppression through negative control of cell population growth mediated by pro-apoptotic signaling.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Membrane Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Klotho Proteins , MAP Kinase Signaling System/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Fibroblast Growth Factor, Type 4/genetics , TOR Serine-Threonine Kinases
15.
Prostate ; 71(15): 1691-700, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21446013

ABSTRACT

BACKGROUND: Despite dramatic positive effects, there is evidence that the androgen receptor (AR) may negatively influence prostate tumor progression. Understanding the AR repressor function and how it is subverted is of particular importance in anti-androgen and AR intervention strategies. METHODS: AR, resident FGFR2IIIb, and ectopic FGFR1 were expressed by transfection in the AR-negative epithelial cell line DTE that predominates in cell culture of AR-positive androgen-responsive model Dunning R3327 rat prostate tumors. Androgen-responsiveness at transcription was measured by a luciferase reporter. Cell population growth rates were assessed by cell counts, DNA synthesis, and expression of cell cycle genes. AR variants (ARVs) were assessed by immunochemistry and nuclease protection of mRNA. RESULTS: Expression of AR inhibited cell population growth of AR-negative DTE cells at the G1-S phase of the cell cycle. Ectopic FGFR1, but not resident FGFR2IIIb abrogated the growth inhibitory effects of AR. Appearance of ARVs was coincident with co-expression of FGFR1 and AR and abrogation of the AR-dependent inhibition of cell growth. CONCLUSIONS: DTE cells may represent non-malignant AR-negative progenitors whose population is restricted by activation of AR in vivo. Ectopic expression of epithelial FGFR1, a common observation in tumors, overrides the inhibition of AR and thus may contribute to evolution of androgen and AR independent tumors. These results are consistent with the notion that some tumor cells are negatively restricted by AR and are unleased by androgen-deprivation or ectopic expression of FGFR1. ARV's may play a role in the bypass of the negative restrictions of AR.


Subject(s)
Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Genetic Variation , Immunohistochemistry , Male , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Protein Isoforms , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
BMC Cell Biol ; 11: 89, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21092184

ABSTRACT

BACKGROUND: Autophagy is a dynamic process during which isolation membranes package substrates to form autophagosomes that are fused with lysosomes to form autolysosomes for degradation. Although it is agreed that the LC3II-associated mature autophagosomes move along microtubular tracks, it is still in dispute if the conversion of LC3I to LC3II before autophagosomes are fully mature and subsequent fusion of mature autophagosomes with lysosomes require microtubules. RESULTS: We use biochemical markers of autophagy and a collection of microtubule interfering reagents to test the question. Results show that interruption of microtubules with either microtubule stabilizing paclitaxel or destabilizing nocodazole similarly impairs the conversion of LC3I to LC3II, but does not block the degradation of LC3II-associated autophagosomes. Acetylation of microtubules renders them resistant to nocodazole treatment. Treatment with vinblastine that causes depolymerization of both non-acetylated and acetylated microtubules results in impairment of both LC3I-LC3II conversion and LC3II-associated autophagosome fusion with lysosomes. CONCLUSIONS: Acetylated microtubules are required for fusion of autophagosomes with lysosomes to form autolysosomes.


Subject(s)
Autophagy , Lysosomes/metabolism , Microtubules/metabolism , Phagosomes/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nocodazole/pharmacology , Paclitaxel/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequestosome-1 Protein , Vinblastine/pharmacology
17.
Mol Carcinog ; 48(6): 553-62, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19009564

ABSTRACT

Fibroblast growth factor (FGF) family signaling mediates cell-to-cell communication in development and organ homeostasis in adults. Of the FGF receptor (FGFR) isotypes, FGFR4 is the sole resident isotype present in mature parenchymal hepatocytes. FGFR1 that is normally associated with activated nonparenchymal cells appears ectopically in hepatoma cells. Ectopic expression and chronic activity of FGFR1 in hepatocytes accelerates diethylnitrosamine (DEN)-initiated hepatocarcinogenesis by driving unrestrained cell proliferation and tumor angiogenesis. Hepatocyte FGFR4 mediates liver's role in systemic cholesterol/bile acid and lipid metabolism and affects proper hepatolobular restoration after damage without effect on cell proliferation. Here we ask whether FGFR4 plays a role in progression of hepatocellular carcinoma (HCC). We report that although spontaneous HCC was not detected in livers of FGFR4-deficient mice, the ablation of FGFR4 accelerated DEN-induced hepatocarcinogenesis. In contrast to FGFR1 that induced a strong mitogenic response and depressed rate of cell death in hepatoma cells, FGFR4 failed to induce a mitogenic response and increased the rate of cell death. FGFR1 but not FGFR4 induced cyclin D1 and repressed p27 expression. Analysis of activation of Erk, JNK, and PI3K-related AKT signaling pathways indicated that in contrast to FGFR1, FGFR4 failed to sustain Erk activation and did not activate AKT. These differences may underlie the opposing effects of FGFR1 and FGFR4. These results suggest that in contrast to ectopic FGFR1 that is a strong promoter of hepatoma, resident FGFR4 that mediates differentiated hepatocyte metabolic functions also serves to suppress hepatoma progression.


Subject(s)
Fibroblast Growth Factor 4/metabolism , Liver Neoplasms, Experimental/prevention & control , Receptor, Fibroblast Growth Factor, Type 4/physiology , Animals , Apoptosis , Base Sequence , Cell Proliferation , DNA Primers , Gene Expression Profiling , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
18.
Anticancer Res ; 28(4B): 2141-6, 2008.
Article in English | MEDLINE | ID: mdl-18751387

ABSTRACT

BACKGROUND: This study was undertaken to investigate the radiosensitizing effects of fibroblast growth factor receptor 2IIIb (FGFR2IIIb) in androgen-independent human prostate carcinoma PC-3 cells devoid of normally resident epithelial cell FGFR2IIIb. MATERIALS AND METHODS: A clonal line of PC-3 cells expressing FGFR2IIIb was established by stable transfection. Clonogenic cell survival, apoptosis and cell cycle distribution with and without gamma-irradiation were then compared between FGFR2IIIb-expressing PC-3 cells and control cells mock-transfected with vector alone. RESULTS: Gamma-irradiation resulted in an increase of clonogenic cell death concurrent with enhanced apoptosis and cell cycle arrest in the G2/M-phase in both transfected and untransfected cells. A quantitative analysis of all three parameters indicated that cells expressing FGFR2IIIb were significantly more sensitive to irradiation than control cells. CONCLUSION: These results indicate that restoration of FGFR2IIIb to PC-3 cells enhances their sensitivity to irradiation through acceleration of apoptosis and cell cycle arrest.


Subject(s)
Prostatic Neoplasms/radiotherapy , Receptor, Fibroblast Growth Factor, Type 2/physiology , Apoptosis/physiology , Apoptosis/radiation effects , Cell Division/physiology , Cell Division/radiation effects , Cell Line, Tumor , G2 Phase/physiology , G2 Phase/radiation effects , Gamma Rays , Humans , Male , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/radiotherapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radiation Tolerance , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Transfection
19.
Cancer Res ; 66(3): 1481-90, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452204

ABSTRACT

Fibroblast growth factor (FGF) signaling mediates cell-to-cell communication in development and organ homeostasis in adults. Of the four FGF receptor (FGFR) tyrosine kinases, only FGFR4 is expressed in mature hepatocytes. Although FGFR1 is expressed by hepatic cell progenitors and adult nonparenchymal cells, ectopic expression is commonly observed in hepatoma cells. Here, we determined whether ectopic FGFR1 is a cause or consequence of hepatocellular carcinoma by targeting a constitutively active human FGFR1 to mouse hepatocytes. Livers of transgenic mice exhibited accelerated regeneration after partial hepatectomy but no signs of neoplastic or preneoplastic abnormalities for up to 18 months. However, in diethylnitrosamine-treated mice, the chronic FGFR1 activity promoted an incidence of 44% adenomas at 4 months and 38% hepatocellular carcinoma at 8 months. No adenoma or hepatocellular carcinoma was observed in diethylnitrosamine-treated wild-type (WT) livers at 4 or 8 months, respectively. At 10 and 12 months, tumor-bearing livers in transgenic mice were twice the size of those in WT animals. Isolated hepatoma cells from the transgenic tumors exhibited a growth advantage in culture. Advanced hepatocellular carcinoma in the transgenic livers exhibited a reduced rate of necrosis. This was accompanied by a mean microvessel density of 2.7 times that of WT tumors and a markedly higher level of vascular endothelial growth factor. In cooperation with an initiator, the persistent activity of ectopic FGFR1 in hepatocytes is a strong promoter of hepatocellular carcinoma by driving cell proliferation at early stages and promoting neoangiogenesis at late stages of progression.


Subject(s)
Hepatocytes/metabolism , Liver Neoplasms, Experimental/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Carcinogens , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA, Neoplasm/biosynthesis , Diethylnitrosamine , Hepatectomy , Hepatocytes/pathology , Humans , Liver/drug effects , Liver/physiology , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/pathology , Liver Regeneration/physiology , MAP Kinase Signaling System , Mice , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
20.
In Vitro Cell Dev Biol Anim ; 54(3): 177-193, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435725

ABSTRACT

Gordon H. Sato, an innovator in mammalian tissue culture and integrated cellular physiology, passed away in 2017. In tribute to Dr. Sato, In Vitro Cellular and Developmental Biology-Animal presents a collection of invited remembrances from six colleagues whose associations with Dr. Sato spanned more than 40 years. Dr. Sato was a past president of the Tissue Culture Association (now the Society for In Vitro Biology), editor-in-chief of In Vitro Cellular and Developmental Biology (1987-1991), and the recipient of the lifetime achievement award from the Society for In Vitro Biology (2002). He was elected to the US National Academy of Sciences in 1984.


Subject(s)
Cell Culture Techniques/history , Mammals/growth & development , Animals , History, 20th Century , History, 21st Century , Humans , United States
SELECTION OF CITATIONS
SEARCH DETAIL