Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Trends Mol Med ; 24(11): 919-930, 2018 11.
Article in English | MEDLINE | ID: mdl-30213702

ABSTRACT

The promise of tissue engineering and regenerative medicine to reduce the burden of disease and improve quality of life are widely acknowledged. Traditional tissue engineering and regenerative medicine approaches rely on generation of tissue constructs in vitro for subsequent transplantation or injection of exogenously manipulated cells into a host. While promising, few such therapies have succeeded in clinical practice. Here, we propose that recent advances in stem cell and developmental biology, immunology, bioengineering, and material sciences, position us to develop a new generation of in vivo regenerative medicine therapies, which we term autotherapies. Autotherapies are strategies based on optimizing endogenous tissue responses and capitalizing on manipulation of stem cell niches and endogenous tissue microenvironments to enhance tissue healing and regeneration.


Subject(s)
Regeneration , Wound Healing , Animals , Cell Lineage/genetics , Cellular Microenvironment , Cellular Reprogramming/genetics , Epigenesis, Genetic , Extracellular Matrix/metabolism , Humans , Stem Cell Niche , Stem Cells/cytology , Stem Cells/metabolism
2.
JCI Insight ; 3(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29415879

ABSTRACT

Several reports have demonstrated that mouse Cx3cr1 signaling promotes monocyte/macrophage survival. In agreement, we previously found that, in a mouse model of systemic candidiasis, genetic deficiency of Cx3cr1 resulted in increased mortality and impaired tissue fungal clearance associated with decreased macrophage survival. We translated this finding by showing that the dysfunctional CX3CR1 variant CX3CR1-M280 was associated with increased risk and worse outcome of human systemic candidiasis. However, the impact of this mutation on human monocyte/macrophage survival is poorly understood. Herein, we hypothesized that CX3CR1-M280 impairs human monocyte survival. We identified WT (CX3CR1-WT/WT), CX3CR1-WT/M280 heterozygous, and CX3CR1-M280/M280 homozygous healthy donors of European descent, and we show that CX3CL1 rescues serum starvation-induced cell death in CX3CR1-WT/WT and CX3CR1-WT/M280 but not in CX3CR1-M280/M280 monocytes. CX3CL1-induced survival of CX3CR1-WT/WT monocytes is mediated via AKT and ERK activation, which are both impaired in CX3CR1-M280/M280 monocytes, associated with decreased blood monocyte counts in CX3CR1-M280/M280 donors at steady state. Instead, CX3CR1-M280/M280 does not affect monocyte CX3CR1 surface expression or innate immune effector functions. Together, we show that homozygocity of the M280 polymorphism in CX3CR1 is a potentially novel population-based genetic factor that influences human monocyte signaling.


Subject(s)
CX3C Chemokine Receptor 1/genetics , Cell Survival/genetics , Monocytes/physiology , Apoptosis/immunology , CX3C Chemokine Receptor 1/metabolism , Cell Culture Techniques , Cell Survival/immunology , Cells, Cultured , Chemokine CX3CL1/immunology , Chemokine CX3CL1/metabolism , Culture Media, Serum-Free , Healthy Volunteers , Homozygote , Humans , Mutation
4.
Sci Signal ; 10(484)2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28634209

ABSTRACT

The ß2-adrenergic receptor (ß2AR) has provided a paradigm to elucidate how G protein-coupled receptors (GPCRs) control intracellular signaling, including the discovery that ß-arrestins, which bind to ligand-activated GPCRs, are central for GPCR function. We used genome editing, conditional gene deletion, and small interfering RNAs (siRNAs) to determine the roles of ß-arrestin 1 (ß-arr1) and ß-arr2 in ß2AR internalization, trafficking, and signaling to ERK. We found that only ß-arr2 was essential for ß2AR internalization. Unexpectedly, ß-arr1 and ß-arr2 and receptor internalization were dispensable for ERK activation. Instead, ß2AR signaled through Gαs and Gßγ subunits through a pathway that involved the tyrosine kinase SRC, the adaptor protein SHC, the guanine nucleotide exchange factor SOS, the small GTPase RAS, and the kinases RAF and MEK, which led to ERK activation. These findings provide a molecular framework for ß2AR signaling through ß-arrestin-independent pathways in key physiological functions and under pathological conditions.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Animals , Endocytosis , GTP Phosphohydrolases/metabolism , HEK293 Cells , Humans , Ligands , Mice , Mice, Knockout , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Transcription Activator-Like Effector Nucleases
5.
Protein Sci ; 23(12): 1708-16, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25220262

ABSTRACT

ARRDC3 is one of six known human α-arrestins, and has been implicated in the downregulation of the ß2-adrenergic receptor (ß2AR). ARRDC3 consists of a two-lobed arrestin fold and a C-terminal tail containing two PPYX motifs. In the current model for receptor downregulation by ARRDC3, the arrestin fold portion is thought to bind the receptor, while the PPXY motifs recruit ubiquitin ligases of the NEDD4 family. Here we report the crystal structures of the N-terminal lobe of human ARRDC3 in two conformations, at 1.73 and 2.8 Å resolution, respectively. The structures reveal a large electropositive region that is capable of binding phosphate ions of crystallization. Residues within the basic patch were shown to be important for binding to ß2AR, similar to the situation with ß-arrestins. This highlights potential parallels in receptor recognition between α- and ß-arrestins.


Subject(s)
Arrestins/chemistry , Arrestins/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Arrestins/isolation & purification , Binding Sites , HEK293 Cells , Humans , Models, Molecular , Protein Conformation , Receptors, Adrenergic, beta-2/isolation & purification
6.
Curr Opin Cell Biol ; 27: 126-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24508914

ABSTRACT

G protein-coupled receptors (GPCRs) play a central role in signal transmission, thereby controlling many facets of cellular function. Overwhelming evidence now implicates GPCRs, G proteins and their downstream signaling targets in cancer initiation and progression, where they can influence aberrant cell growth and survival, largely through activation of AKT/mTOR, MAPKs, and Hippo signaling pathways. GPCRs also play critical roles in the invasion and metastasis of cancer cells via activation of Rho GTPases and cytoskeletal changes, and angiogenesis to supply the tumor with nutrients and provide routes for metastasis. Lastly, GPCRs contribute to the establishment and maintenance of a permissive tumor microenvironment. Understanding GPCR involvement in cancer malignancy may help identify novel therapeutic opportunities for cancer prevention and treatment.


Subject(s)
GTP-Binding Proteins/metabolism , Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Cell Proliferation , Cell Survival , Cytoskeleton/metabolism , GTP-Binding Proteins/genetics , Humans , MAP Kinase Signaling System , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk , Receptors, G-Protein-Coupled/genetics , Second Messenger Systems , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Viral Proteins/metabolism , rho GTP-Binding Proteins/metabolism
7.
J Biol Chem ; 289(8): 4743-52, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24379409

ABSTRACT

After protracted stimulation, the ß2-adrenergic receptor and many other G-protein-coupled receptors are ubiquitinated and down-regulated. Arrestin-related domain-containing protein-3 (ARRDC3) has been proposed to recruit the ubiquitin ligase Nedd4 to the ß2-adrenergic receptor. ARRDC3 contains two PPXY motifs that could potentially interact with any of the four WW domains of Nedd4. Here we dissect the interaction determinants. ARRDC3 PPXY-Nedd4 WW dissociation constants vary from unmeasurable to Kd = 3 µM for the third WW domain of Nedd4 binding to the first PPXY motif of ARRDC3. Structures of the uncomplexed and PPXY1-bound WW3 domain were determined at 1.1 and 1.7 Å resolution. The structures revealed conformational changes upon binding and the hydrogen bonding network in exquisite detail. Tight packing of ARRDC3 Val-352', part of a 310 helix at the C terminus of PPXY1, is important for high affinity binding to WW3. Although no single WW domain is strictly essential for the binding of Nedd4 and ARRDC3 expressed in HEK293 cells, high affinity binding of full-length ARRDC3 and Nedd4 is driven by the avid interaction of both PPXY motifs with either the WW2-WW3 or WW3-WW4 combinations, with Kd values as low as 300 nM.


Subject(s)
Arrestins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Arrestins/chemistry , Calorimetry , Crystallography, X-Ray , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Models, Molecular , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Nedd4 Ubiquitin Protein Ligases , Proline/analogs & derivatives , Proline/metabolism , Protein Binding , Protein Structure, Tertiary , Ubiquitin-Protein Ligases/chemistry
8.
Nat Rev Cancer ; 13(6): 412-24, 2013 06.
Article in English | MEDLINE | ID: mdl-23640210

ABSTRACT

Aberrant expression and activity of G proteins and G-protein-coupled receptors (GPCRs) are frequently associated with tumorigenesis. Deep sequencing studies show that 4.2% of tumours carry activating mutations in GNAS (encoding Gαs), and that oncogenic activating mutations in genes encoding Gαq family members (GNAQ or GNA11) are present in ~66% and ~6% of melanomas arising in the eye and skin, respectively. Furthermore, nearly 20% of human tumours harbour mutations in GPCRs. Many human cancer-associated viruses also express constitutively active viral GPCRs. These studies indicate that G proteins, GPCRs and their linked signalling circuitry represent novel therapeutic targets for cancer prevention and treatment.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Neoplasms/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , Gene Expression , Gene Frequency , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mutation , Neoplasms/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
9.
Mol Med ; 18: 19-28, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-21979753

ABSTRACT

We have previously shown that sorafenib, a multikinase inhibitor, exhibits cytotoxic effects on chronic lymphocytic leukemia (CLL) cells. Because the cellular microenvironment can protect CLL cells from drug-induced apoptosis, it is important to evaluate the effect of novel drugs in this context. Here we characterized the in vitro cytotoxic effects of sorafenib on CLL cells and the underlying mechanism in the presence of marrow stromal cells (MSCs) and nurselike cells (NLCs). One single dose of 10 µmol/L or the repeated addition of 1 µmol/L sorafenib caused caspase-dependent apoptosis and reduced levels of phosphorylated B-RAF, C-RAF, extracellular signal-regulated kinase (ERK), signal transducer and activator of transcription 3 (STAT3) and myeloid cell leukemia sequence 1 (Mcl-1) in CLL cells in the presence of the microenvironment. We show that the RAF/mitogen-activated protein kinase kinase (MEK)/ERK pathway can modulate Mcl-1 expression and contribute to CLL cell viability, thereby associating so-rafenib cytotoxicity to its impact on RAF and Mcl-1. To evaluate if the other targets of sorafenib can affect CLL cell viability and contribute to sorafenib-mediated cytotoxicity, we tested the sensitivity of CLL cells to several kinase inhibitors specific for these targets. Our data show that RAF and vascular endothelial growth factor receptor (VEGFR) but not KIT, platelet-derived growth factor receptor (PDGFR) and FMS-like tyrosine kinase 3 (FLT3) are critical for CLL cell viability. Taken together, our data suggest that sorafenib exerts its cytotoxic effect likely via inhibition of the VEGFR and RAF/MEK/ERK pathways, both of which can modulate Mcl-1 expression in CLL cells. Furthermore, sorafenib induced apoptosis of CLL cells from fludarabine refractory patients in the presence of NLCs or MSCs. Our results warrant further clinical exploration of sorafenib in CLL.


Subject(s)
Apoptosis/drug effects , Benzenesulfonates/pharmacology , Cell Survival/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridines/pharmacology , raf Kinases/metabolism , Cells, Cultured , Humans , Immunoblotting , Leukemia, Lymphocytic, Chronic, B-Cell , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Sorafenib , Tumor Cells, Cultured
10.
Blood ; 117(3): 882-9, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21079155

ABSTRACT

The chemokine CXCL12, via its receptor CXCR4, promotes increased survival of chronic lymphocytic leukemia (CLL) B cells that express high levels of ζ-chain-associated protein (ZAP-70), a receptor tyrosine kinase associated with aggressive disease. In this study, we investigated the underlying molecular mechanisms governing this effect. Although significant differences in the expression or turnover of CXCR4 were not observed between ZAP-70(+) and ZAP-70(-) cell samples, CXCL12 induced greater intracellular Ca(2+) flux and stronger and more prolonged phosphorylation of extracellular signal-regulated kinase (ERK) and mitogen-activated protein kinase/ERK kinase (MEK) in the ZAP-70(+) CLL cells. The CXCL12-induced phosphorylation of ERK and MEK in ZAP-70(+) CLL cells was blocked by sorafenib, a small molecule inhibitor of RAF. Furthermore, ZAP-70(+) CLL cells were more sensitive than ZAP-70(-) CLL cells to the cytotoxic effects of sorafenib in vitro at concentrations that can readily be achieved in vivo. The data suggest that ZAP-70(+) CLL cells may be more responsive to survival factors, like CXCL12, that are elaborated by the leukemia microenvironment, and this sensitivity could be exploited for the development of new treatments for patients with this disease. Moreover, sorafenib may have clinical activity for patients with CLL, particularly those with ZAP-70(+) CLL.


Subject(s)
Benzenesulfonates/pharmacology , Chemokine CXCL12/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , raf Kinases/metabolism , Apoptosis/drug effects , Calcium/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Humans , Immunoblotting , Intracellular Space/drug effects , Intracellular Space/metabolism , Jurkat Cells , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mitogen-Activated Protein Kinases/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, CXCR4/metabolism , Sorafenib , Time Factors , Tumor Cells, Cultured , ZAP-70 Protein-Tyrosine Kinase/metabolism , raf Kinases/antagonists & inhibitors
11.
Expert Opin Drug Discov ; 5(11): 1109-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21132095

ABSTRACT

IMPORTANCE OF THE FIELD: Chemokine receptors are most noted for their role in cell migration. However, inappropriate utilization or regulation of these receptors is implicated in many inflammatory diseases, cancer and HIV, making them important drug targets. AREAS COVERED IN THIS REVIEW: Allostery, oligomerization and ligand bias are presented as they pertain to chemokine receptors and their associated pathologies.Specific examples of each are described from the recent literature and their implications are discussed in terms of drug discovery efforts targeting chemokine receptors. WHAT THE READER WILL GAIN: Insight into the expanding view of the multitude of pharmacological variables that need to be considered or that may be exploited in chemokine receptor drug discovery. TAKE HOME MESSAGE: Since 2007, two drugs targeting chemokine receptors have been approved by the FDA, Maraviroc for preventing HIV infection and Mozobil™ for hematopoietic stem cell mobilization. While these successes permit optimism for chemokine receptors as drug targets, only recently has the complexity of this system begun to be appreciated. The concepts of allosteric inhibitors, biased ligands and functional selectivity raise the possibility that drugs with precisely-defined properties can be developed. Other complexities such as receptor oligomerization and tissue-specific functional states of receptors also offer opportunities for increased target and response specificity, although it will be more challenging to translate these ideas into approved therapeutics compared to traditional approaches.


Subject(s)
Drug Discovery/methods , Receptors, Chemokine/drug effects , Animals , Drug Approval , Humans , Molecular Conformation , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/drug effects , Small Molecule Libraries
12.
PLoS One ; 5(7): e11716, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20661426

ABSTRACT

BACKGROUND: Chronic Lymphocytic Leukemia (CLL) pathogenesis has been linked to the prolonged survival and/or apoptotic resistance of leukemic B cells in vivo, and is thought to be due to enhanced survival signaling responses to environmental factors that protect CLL cells from spontaneous and chemotherapy-induced death. Although normally associated with cell migration, the chemokine, CXCL12, is one of the factors known to support the survival of CLL cells. Thus, the signaling pathways activated by CXCL12 and its receptor, CXCR4, were investigated as components of these pathways and may represent targets that if inhibited, could render resistant CLL cells more susceptible to chemotherapy. METHODOLOGY/PRINCIPAL FINDINGS: To determine the downstream signaling targets that contribute to the survival effects of CXCL12 in CLL, we took a phosphoproteomics approach to identify and compare phosphopeptides in unstimulated and CXCL12-stimulated primary CLL cells. While some of the survival pathways activated by CXCL12 in CLL are known, including Akt and ERK1/2, this approach enabled the identification of additional signaling targets and novel phosphoproteins that could have implications in CLL disease and therapy. In addition to the phosphoproteomics results, we provide evidence from western blot validation that the tumor suppressor, programmed cell death factor 4 (PDCD4), is a previously unidentified phosphorylation target of CXCL12 signaling in all CLL cells probed. Additionally, heat shock protein 27 (HSP27), which mediates anti-apoptotic signaling and has previously been linked to chemotherapeutic resistance, was detected in a subset (approximately 25%) of CLL patients cells examined. CONCLUSIONS/SIGNIFICANCE: Since PDCD4 and HSP27 have previously been associated with cancer and regulation of cell growth and apoptosis, these proteins may have novel implications in CLL cell survival and represent potential therapeutic targets. PDCD4 also represents a previously unknown signaling target of chemokine receptors; therefore, these observations increase our understanding of alternative pathways to migration that may be activated or inhibited by chemokines in the context of cancer cell survival.


Subject(s)
Chemokine CXCL12/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Proteomics/methods , Receptors, CXCR4/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12/genetics , Flow Cytometry , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mass Spectrometry , Models, Biological , Phosphorylation , Receptors, CXCR4/genetics , Signal Transduction/genetics
13.
Methods Enzymol ; 460: 331-46, 2009.
Article in English | MEDLINE | ID: mdl-19446733

ABSTRACT

Chemokines induce a number of intracellular signaling pathways by activating second messengers (e.g. calcium) and phosphorylation cascades in order to mediate a myriad of functions including cell migration, survival and proliferation. Although there is some degree of overlap in chemokine receptor-mediated pathway activation, different chemokines will often elicit distinct signaling events. Factors such as cell type, receptor expression levels, G protein availability, and disease state will also influence the signaling response from chemokine-induced receptor activation. Improvements in mass spectrometry, enrichment strategies, and database search programs for identifying phosphopeptides have made phosphoproteomics an accessible biological tool for studying chemokine-induced phosphorylation cascades. Although signaling pathways involved in chemokine-mediated migration have been fairly well characterized, less is known regarding other signaling cascades elicited by chemokines (e.g. to induce proliferation) or the potential for distinct pathway activation in a disease state such as cancer. CXCL12(SDF-1)/CXCR4 signaling has been shown to play an important role in the survival of chronic lymphocytic leukemia (CLL) cells, and thus provides a good system for exploring chemokine signaling, particularly in the interest of survival pathway activation. In this chapter, we describe the use of immobilized metal affinity chromatography (IMAC) phosphopeptide enrichment followed by reversed-phase liquid chromatography and tandem mass spectrometry (LC-MS/MS) analysis for exploring CXCL12-mediated signaling in human CLL patient cells.


Subject(s)
Chemokine CXCL12/metabolism , Proteomics/methods , Receptors, CXCR4/metabolism , Signal Transduction/physiology , Chromatography, Affinity , Chromatography, Liquid , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Phosphorylation , Signal Transduction/genetics , Tandem Mass Spectrometry
14.
Biochem J ; 409(3): 635-49, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18177271

ABSTRACT

Inappropriate chemokine/receptor expression or regulation is linked to many diseases, especially those characterized by an excessive cellular infiltrate, such as rheumatoid arthritis and other inflammatory disorders. There is now overwhelming evidence that chemokines are also involved in the progression of cancer, where they function in several capacities. First, specific chemokine-receptor pairs are involved in tumour metastasis. This is not surprising, in view of their role as chemoattractants in cell migration. Secondly, chemokines help to shape the tumour microenvironment, often in favour of tumour growth and metastasis, by recruitment of leucocytes and activation of pro-inflammatory mediators. Emerging evidence suggests that chemokine receptor signalling also contributes to survival and proliferation, which may be particularly important for metastasized cells to adapt to foreign environments. However, there is considerable diversity and complexity in the chemokine network, both at the chemokine/receptor level and in the downstream signalling pathways they couple into, which may be key to a better understanding of how and why particular chemokines contribute to cancer growth and metastasis. Further investigation into these areas may identify targets that, if inhibited, could render cancer cells more susceptible to chemotherapy.


Subject(s)
Cell Movement , Chemokines/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Animals , Cell Communication/immunology , Cell Movement/immunology , Chemokines/chemistry , Chemokines/immunology , Humans , Neoplasm Metastasis/pathology , Neoplasms/immunology , Signal Transduction/immunology
15.
Am J Physiol Renal Physiol ; 293(3): F846-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17596530

ABSTRACT

During systemic acidosis, renal proximal tubular cells exhibit enhanced rates of bicarbonate and ammonium ion synthesis and undergo extensive hypertrophy. The former adaptations are accomplished, in part, by increased expression of glutaminase (GA). LLC-PK(1)-FBPase+ cells, a gluconeogenic line of porcine kidney cells, exhibit a rapid activation of the ERK1/2 and p38 MAPK pathways and a two- to threefold increase in GA mRNA when transferred to acidic medium (pH 6.9). Transforming growth factor-beta (TGF-beta), a potent activator of MAPK and Smad signaling cascades, also causes extensive renal hypertrophy. Thus the potential role of TGF-beta in the renal response to metabolic acidosis was investigated. Western blot analyses established that in LLC-PK(1)-FBPase+ cells, TGF-beta activated the ERK1/2, p38 MAPK, and Smad1/5/8 pathways, but not the JNK and Smad2/3 pathways. Addition of TGF-beta to cells cultured in normal medium (pH 7.4) produced a steady increase in GA mRNA, resulting in a twofold induction after 18 h. Western blot analysis indicated that treatment with either TGF-beta or acidic medium resulted in an increased level of fibronectin. However, the effects of the two treatments on both GA mRNA and fibronectin levels occurred with different time courses and were additive. In addition, the rates of ammonia production were decreased slightly by addition of TGF-beta. Finally, a GA-luciferase reporter construct, which is activated 3.5-fold by treatment with acidic medium, is not affected by TGF-beta. Therefore, TGF-beta and metabolic acidosis activate some of the same signaling pathways in LLC-PK(1)-FBPase+ cells, but produce separate effects on GA expression.


Subject(s)
Glutaminase/genetics , Glutaminase/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad Proteins/metabolism , Swine , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Biol Chem ; 281(5): 2982-8, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16319064

ABSTRACT

Metabolic acidosis is partially compensated by a pronounced increase in renal catabolism of glutamine. This adaptive response is sustained, in part, through increased expression of phosphoenolpyruvate carboxykinase (PEPCK). Previous inhibitor studies suggested that the pH-responsive increase in PEPCK mRNA in LLC-PK1-FBPase+ cells is mediated by a p38 mitogen-activated protein kinase (MAPK). These cells express high levels of the upstream kinase MAPK kinase (MKK) 3 but relatively low levels of the alternative upstream kinase MKK6. To firmly establish the role of the p38 MAPK signaling pathway, clonal lines of LLC-PK1-FBPase+ cells that express constitutively active (ca) and dominant negative (dn) forms of MKK3 and MKK6 from a tetracycline-responsive promoter were developed. Western blot analyses confirmed that 0.5 microg/ml doxycycline was sufficient to block transcription and that removal of doxycycline led to pronounced and sustained expression of the caMKKs and dnMKKs. Expression of caMKK6 (but not caMKK3) caused an increase in phosphorylation of p38 MAPK and an increase in the level of PEPCK mRNA that closely mimicked the effect of treatment with acidic medium (pH 6.9, 10 mm HCO3-). Only caMKK6 activated transcription of a PEPCK-luciferase reporter construct. Co-expression of both dnMKKs blocked the increases in phosphorylation of p38 MAPK and PEPCK mRNA. The latter effect closely mimicked that of the p38 MAPK inhibitor SB203580. The expression of either dnMKK3 or dnMKK6 was less effective than co-expression of both dnMKKs. Thus, the pH-responsive increase in PEPCK mRNA in the kidney is mediated by the p38 MAPK signaling pathway and involves activation of MKK3 and/or MKK6.


Subject(s)
MAP Kinase Kinase 3/physiology , MAP Kinase Kinase 6/pharmacokinetics , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Acidosis , Animals , Cell Line , Chick Embryo , Hydrogen-Ion Concentration , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/metabolism , MAP Kinase Signaling System , Phosphorylation , RNA, Messenger , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...