Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Med ; 21(9): 1085-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26236992

ABSTRACT

Myostatin (also known as growth and differentiation factor 8) is a secreted member of the transforming growth factor-ß (TGF-ß) family that is mainly expressed in skeletal muscle, which is also its primary target tissue. Deletion of the myostatin gene (Mstn) in mice leads to muscle hypertrophy, and animal studies support the concept that myostatin is a negative regulator of muscle growth and regeneration. However, myostatin deficiency also increases bone formation, mainly through loading-associated effects on bone. Here we report a previously unknown direct role for myostatin in osteoclastogenesis and in the progressive loss of articular bone in rheumatoid arthritis (RA). We demonstrate that myostatin is highly expressed in the synovial tissues of RA subjects and of human tumor necrosis factor (TNF)-α transgenic (hTNFtg) mice, a model for human RA. Myostatin strongly accelerates receptor activator of nuclear factor κB ligand (RANKL)-mediated osteoclast formation in vitro through transcription factor SMAD2-dependent regulation of nuclear factor of activated T-cells (NFATC1). Myostatin deficiency or antibody-mediated inhibition leads to an amelioration of arthritis severity in hTNFtg mice, chiefly reflected by less bone destruction. Consistent with these effects in hTNFtg mice, the lack of myostatin leads to increased grip strength and less bone erosion in the K/BxN serum-induced arthritis model in mice. The results strongly suggest that myostatin is a potent therapeutic target for interfering with osteoclast formation and joint destruction in RA.


Subject(s)
Arthritis, Rheumatoid/therapy , Cell Differentiation , Myostatin/physiology , Osteoclasts/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Myostatin/antagonists & inhibitors , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , Osteogenesis , RANK Ligand/pharmacology
2.
Arthritis Res Ther ; 17: 12, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25600705

ABSTRACT

INTRODUCTION: Inflammatory destructive arthritis, like rheumatoid arthritis (RA), is characterized by invasion of synovial fibroblasts (SF) into the articular cartilage and erosion of the underlying bone, leading to progressive joint destruction. Because fibroblast activation protein alpha (FAP) has been associated with cell migration and cell invasiveness, we studied the function of FAP in joint destruction in RA. METHODS: Expression of FAP in synovial tissues and fibroblasts from patients with osteoarthritis (OA) and RA as well as from wild-type and arthritic mice was evaluated by immunohistochemistry, fluorescence microscopy and polymerase chain reaction (PCR). Fibroblast adhesion and migration capacity was assessed using cartilage attachment assays and wound-healing assays, respectively. For in vivo studies, FAP-deficient mice were crossed into the human tumor necrosis factor transgenic mice (hTNFtg), which develop a chronic inflammatory arthritis. Beside clinical assessment, inflammation, cartilage damage, and bone erosion were evaluated by histomorphometric analyses. RESULTS: RA synovial tissues demonstrated high expression of FAP whereas in OA samples only marginal expression was detectable. Consistently, a higher expression was detected in arthritis SF compared to non-arthritis OA SF in vitro. FAP-deficiency in hTNFtg mice led to less cartilage degradation despite unaltered inflammation and bone erosion. Accordingly, FAP(-/-) hTNFtg SF demonstrated a lower cartilage adhesion capacity compared to hTNFtg SF in vitro. CONCLUSIONS: These data point to a so far unknown role of FAP in the attachment of SF to cartilage, promoting proteoglycan loss and subsequently cartilage degradation in chronic inflammatory arthritis.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Gelatinases/deficiency , Membrane Proteins/deficiency , Serine Endopeptidases/deficiency , Animals , Arthritis, Rheumatoid/prevention & control , Endopeptidases , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proteoglycans/deficiency
3.
Ann Rheum Dis ; 74(12): 2216-23, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25125695

ABSTRACT

OBJECTIVE: We analysed the role of the adaptor molecule four-and-a-half Lin11, Isl-1 & Mec-3 (LIM) domain protein 2 (FHL2) in the activation of fibroblast-like synoviocytes in human rheumatoid arthritis (RA) and tumour necrosis factor α (TNFα)-dependent animal models of the disease. METHODS: Synovial tissues of patients with RA and osteoarthritis (OA) as well as hind paw sections from arthritic human TNFα transgenic (hTNFtg) mice and synovial fibroblasts from these were analysed. The effects of cytokines on the expression of FHL2 and disease-relevant matrixmetalloproteases (MMPs) were determined. Analyses of human tissue specimens from patients treated with anti-TNFα as well as anti-TNFα treatment of hTNFtg mice were performed to substantiate the TNFα effects on FHL2 levels. FHL2(-/-) mice and hTNFtg mice (with constitutive or inducible transgene expression) were crossbred to generate TNFα overexpressing FHL2-deficient animals. Signalling pathways were analysed in cells from these mice and in human cells after knock down of FHL2 by western blot. RESULTS: FHL2 levels were higher in RA than in OA and in hTNFtg than in wild-type mice. Surprisingly, while transforming growth factor (TGF)ß-induced FHL2 expression, TNFα suppressed FHL2. In vivo, anti-TNFα treatment led to higher FHL2 levels both in RA patients and hTNFtg mice. The loss of FHL2 increased joint destruction in hTNFtg mice, which was accompanied by elevated MMP-13. In vitro, TNFα-mediated MMP-13 was significantly higher in FHL2(-/-) cells and after knock down of FHL2, which was caused by prolonged p38 MAPK activation. CONCLUSIONS: These data suggest that FHL2 serves as a protective factor and that, rather than promoting the pathology, the upregulation of FHL2 in RA occurs in frame of a regenerative attempt.


Subject(s)
DNA/genetics , Gene Expression Regulation , LIM-Homeodomain Proteins/genetics , Muscle Proteins/genetics , Osteoarthritis/genetics , Synovial Membrane/metabolism , Transcription Factors/genetics , Animals , Cells, Cultured , Chronic Disease , Humans , Immunoblotting , LIM-Homeodomain Proteins/biosynthesis , Mice , Mice, Transgenic , Muscle Proteins/biosynthesis , Osteoarthritis/metabolism , Osteoarthritis/pathology , Real-Time Polymerase Chain Reaction , Signal Transduction , Synovial Membrane/pathology , Transcription Factors/biosynthesis
4.
PLoS One ; 8(7): e70709, 2013.
Article in English | MEDLINE | ID: mdl-23894681

ABSTRACT

BACKGROUND: The matrix metalloproteinases (MMPs) and their endogenous regulators, the tissue inhibitor of metalloproteinases (TIMPs 1-4) are responsible for the physiological remodeling of the extracellular matrix (ECM). Among all TIMPs, TIMP3 appears to play a unique role since TIMP3 is a secreted protein and, unlike the other TIMP family members, is tightly bound to the ECM. Moreover TIMP3 has been shown to be able to induce apoptotic cell death. As little is known about the underlying mechanisms, we set out to investigate the pro-apoptotic effect of TIMP3 in human mesenchymal cells. METHODOLOGY/PRINCIPAL FINDINGS: Lentiviral overexpression of TIMP3 in mesenchymal cells led to a strong dose-dependent induction of ligand-independent apoptosis as reflected by a five-fold increase in caspase 3 and 7 activity compared to control (pLenti6/V5-GW/lacZ) or uninfected cells, whereas exogenous TIMP3 failed to induce apoptosis. Concordantly, increased cleavage of death substrate PARP and the caspases 3 and 7 was observed in TIMP3 overexpressing cultures. Notably, activation of caspase-8 but not caspase-9 was observed in TIMP3-overexpressing cells, indicating a death receptor-dependent mechanism. Moreover, overexpression of TIMP3 led to a further induction of apoptosis after stimulation with TNF-alpha, FasL and TRAIL. Most interestingly, TIMP3-overexpression was associated with a decrease in phosphorylation of cRaf, extracellular signal-regulated protein kinase (Erk1/2), ribosomal S6 kinase (RSK1) and Akt and serum deprivation of TIMP3-overexpressing cells resulted in a distinct enhancement of apoptosis, pointing to an impaired signaling of serum-derived survival factors. Finally, heparinase treatment of heparan sulfate proteoglycans led to the release of TIMP3 from the surface of overexpressing cells and to a significant decrease in apoptosis indicating that the binding of TIMP3 is necessary for apoptosis induction. CONCLUSION: The results demonstrate that exclusively cell surface-bound endogenous TIMP3 induces apoptosis in mesenchymal Cal78 cells through ligand-independent activation of death receptor signaling and blockade of survival signaling pathways.


Subject(s)
Tissue Inhibitor of Metalloproteinase-3/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line , Cell Line, Tumor , Dogs , Heparan Sulfate Proteoglycans/metabolism , Heparin Lyase/metabolism , Humans , Lentivirus/genetics , Mesoderm/metabolism , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinase-3/genetics
5.
Ann Rheum Dis ; 72(11): 1874-81, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23417988

ABSTRACT

OBJECTIVE: Based on previous data that have linked the small ubiquitin-like modifier-1 (SUMO-1) to the pathogenesis of rheumatoid arthritis (RA), we have investigated the expression of the highly homologous SUMO family members SUMO-2/3 in human RA and in the human tumour necrosis factor α transgenic (hTNFtg) mouse model of RA and studied their role in regulating disease specific matrixmetalloproteinases (MMPs). METHODS: Synovial tissue was obtained from RA and osteoarthritis (OA) patients and used for histological analyses as well as for the isolation of synovial fibroblasts (SFs). The expression of SUMO-2/3 in RA and OA patients as well as in hTNFtg and wild type mice was studied by PCR, western blot and immunostaining. SUMO-2/3 was knocked down using small interfering RNA in SFs, and TNF-α induced MMP production was determined by ELISA. Activation of nuclear factor-κB (NF-κB) was determined by a luciferase activity assay and a transcription factor assay in the presence of the NF-κB inhibitor BAY 11-7082. RESULTS: Expression of SUMO-2 and to a lesser extent of SUMO-3 was higher in RA tissues and RASFs compared with OA controls. Similarly, there was increased expression of SUMO-2 in the synovium and in SFs of hTNFtg mice compared with wild type animals. In vitro, the expression of SUMO-2 but not of SUMO-3 was induced by TNF-α. The knockdown of SUMO-2/3 significantly increased the TNF-α and interleukin (IL)-1ß induced expression of MMP-3 and MMP-13, accompanied by increased NF-κB activity. Induction of MMP-3 and MMP-13 was inhibited by blockade of the NF-κB pathway. TNF-α and IL-1ß mediated MMP-1 expression was not regulated by SUMO-2/3. CONCLUSIONS: Collectively, we show that despite their high homology, SUMO-2/3 are differentially regulated by TNF-α and selectively control TNF-α mediated MMP expression via the NF-κB pathway. Therefore, we hypothesise that SUMO-2 contributes to the specific activation of RASF.


Subject(s)
Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , NF-kappa B/physiology , Small Ubiquitin-Related Modifier Proteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Humans , Mice , Mice, Transgenic , Osteoarthritis/metabolism , Signal Transduction , Synovial Membrane/cytology , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/genetics , Ubiquitins/physiology
6.
Nat Med ; 15(12): 1414-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19898488

ABSTRACT

Active rheumatoid arthritis originates from few joints but subsequently affects the majority of joints. Thus far, the pathways of the progression of the disease are largely unknown. As rheumatoid arthritis synovial fibroblasts (RASFs) which can be found in RA synovium are key players in joint destruction and are able to migrate in vitro, we evaluated the potential of RASFs to spread the disease in vivo. To simulate the primary joint of origin, we implanted healthy human cartilage together with RASFs subcutaneously into severe combined immunodeficient (SCID) mice. At the contralateral flank, we implanted healthy cartilage without cells. RASFs showed an active movement to the naive cartilage via the vasculature independent of the site of application of RASFs into the SCID mouse, leading to a marked destruction of the target cartilage. These findings support the hypothesis that the characteristic clinical phenomenon of destructive arthritis spreading between joints is mediated, at least in part, by the transmigration of activated RASFs.


Subject(s)
Arthritis, Rheumatoid/pathology , Synovial Membrane/pathology , Animals , Cartilage/pathology , Cartilage/physiopathology , Cell Adhesion , Cell Movement , Disease Progression , Extracellular Matrix , Fibroblasts/pathology , Humans , Mice , Mice, SCID , Synovial Membrane/physiopathology , Transplantation, Heterologous
7.
FASEB J ; 23(12): 4288-98, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19734303

ABSTRACT

The gamma isoform of phosphoinositide 3-kinase (PI3Kgamma) has been viewed as restricted to leukocytes mediating the regulation of chemokine-induced migration and recruitment of neutrophils, monocytes, and macrophages. In line with the observation that PI3Kgamma-deficient mice display defects in adaptive immunity, inhibition of PI3Kgamma reduces synovial inflammation in the collagen-induced arthritis mouse model of inflammatory arthritis [rheumatoid arthritis (RA)], which has been attributed to reduced influx of inflammatory cells. Challenging the concept of leukocyte-restricted PI3Kgamma function, we report here a novel, nonredundant function of PI3Kgamma as an important regulator of fibroblast-induced cartilage destruction during chronic destructive arthritis. We show that in human tumor necrosis factor transgenic mice, the loss of PI3Kgamma leads to a milder inflammatory arthritis. Interestingly, PI3Kgamma deficiency does not alter the recruitment of inflammatory cells, but significantly reduces cartilage damage through reduced expression of matrix metalloproteinases in fibroblasts and chondrocytes. In vitro analyses demonstrate that the decreased invasiveness of fibroblasts is mediated by reduced phosphorylation of Akt and extracellular signal-regulated kinase. Using a PI3Kgamma specific inhibitor, these data are confirmed in human synovial fibroblasts from patients with RA who exhibit a disease-specific up-regulation of PI3Kgamma. Our data indicate that in addition to mediating the recruitment of inflammatory cells, PI3Kgamma is an important regulator of fibroblast-mediated joint destruction in RA and suggest that specific inhibitors of PI3Kgamma will interfere with the activation of RA synovial fibroblasts and reduce cartilage destruction in RA.


Subject(s)
Arthritis/metabolism , Cartilage/pathology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Arthritis/genetics , Arthritis/pathology , Arthritis, Rheumatoid/metabolism , Chondrocytes/metabolism , Class Ib Phosphatidylinositol 3-Kinase , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression/physiology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Metalloproteases/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Synovial Membrane/cytology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
8.
Arthritis Rheum ; 60(9): 2606-11, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19714628

ABSTRACT

OBJECTIVE: Synovial fibroblasts (SFs) contribute to several aspects of the pathogenesis of rheumatoid arthritis (RA) and have been implicated most prominently in the progressive destruction of articular cartilage. Targeting the invasive phenotype of RASFs has therefore gained increasing attention, but the precise measurement of their invasive capacity and the evaluation of potential treatment effects constitute a challenge that needs to be addressed. This study used a novel in vitro invasion assay based on the breakdown of transepithelial electrical resistance to determine the course of fibroblast invasion into extracellular matrix. METHODS: A matrix-associated transepithelial resistance invasion (MATRIN) assay was used to assess SFs from patients with RA in comparison with SFs from patients with osteoarthritis (OA). The SFs were grown on a commercially available collagen mix that was placed onto the upper side of a Transwell polycarbonate membrane. In addition, freshly isolated cartilage extracts were studied to assess the conditions in vivo. Under this membrane, a monolayer of MDCK-C7 cells was seeded to create a high electrical resistance. RESULTS: Invasion of fibroblasts into the matrix affected the integrity of the MDCK-C7 monolayer and led to a measurable decrease and subsequent breakdown of electrical resistance. Unlike in the assay with OASFs, which did not achieve a breakdown of resistance up to 72 hours, RASFs exhibited a pronounced invasiveness in this assay, with a 50% breakdown after 42 hours. Treatment of fibroblasts with either a matrix metalloproteinase inhibitor or antibodies against beta1 integrin significantly reduced the invasiveness of RASFs. CONCLUSION: The MATRIN assay is a valuable and sensitive biologic assay system that can be used to determine precisely the invasive potential of RASFs in vitro, and thus would be suitable for screening anti-invasion compounds.


Subject(s)
Arthritis, Rheumatoid/pathology , Biological Assay/methods , Cell Movement/physiology , Extracellular Matrix/pathology , Fibroblasts/pathology , Osteoarthritis/pathology , Synovial Membrane/pathology , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Communication/physiology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cells, Cultured , Dogs , Electric Impedance , Humans , Integrin beta1/immunology , Kidney/cytology , Melanoma/pathology , Metalloproteases/antagonists & inhibitors , Skin Neoplasms/pathology
9.
Arthritis Res Ther ; 11(1): R16, 2009.
Article in English | MEDLINE | ID: mdl-19196465

ABSTRACT

INTRODUCTION: The rheumatoid arthritis (RA) synovium is characterised by the presence of an aggressive population of activated synovial fibroblasts (RASFs) that are prominently involved in the destruction of articular cartilage and bone. Accumulating evidence suggests that RASFs are relatively resistant to Fas-ligand (FasL)-induced apoptosis, but the data concerning tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) have been conflicting. Here, we hypothesise that the susceptibility of RASFs to receptor-mediated apoptosis depends on the proliferation status of these cells and therefore analysed the cell cycle dependency of FasL- and TRAIL-induced programmed cell death of RASFs in vitro. METHODS: Synovial fibroblasts were isolated from patients with RA by enzymatic digestion and cultured under standard conditions. Cell cycle analysis was performed using flow cytometry and staining with propidium iodide. RASFs were synchronised or arrested in various phases of the cell cycle with 0.5 mM hydroxyurea or 2.5 microg/ml nocodazol and with foetal calf serum-free insulin-transferrin-sodium selenite supplemented medium. Apoptosis was induced by stimulation with 100 ng/ml FasL or 100 ng/ml TRAIL over 18 hours. The apoptotic response was measured using the Apo-ONE Homogenous Caspase-3/7 Assay (Promega GmbH, Mannheim, Germany) and the Cell Death Detection (ELISAPlus) (enzyme-linked immunosorbent assay) (Roche Diagnostics GmbH, Mannheim, Germany). Staurosporin-treated cells (1 microg/ml) served as a positive control. Expression of Fas and TRAIL receptors (TRAILR1-4) was determined by fluorescence-activated cell sorting analysis. RESULTS: Freshly isolated RASFs showed only low proliferation in vitro, and the rate decreased further over time, particularly when RASFs became confluent. RASFs expressed Fas, TRAIL receptor-1, and TRAIL receptor-2, and the expression levels were independent of the cell cycle. However, the proliferation rate significantly influenced the susceptibility to FasL- and TRAIL-induced apoptosis. Specifically, proliferating RASFs were less sensitive to FasL- and TRAIL-induced apoptosis than RASFs with a decreased proliferation rate. Furthermore, RASFs that were synchronised in S phase or G2/M phase were less sensitive to TRAIL-induced apoptosis than synchronised RASFs in G0/G1 phase. CONCLUSIONS: Our data indicate that the susceptibility of RASFs to FasL- and TRAIL-induced apoptosis depends on the cell cycle. These results may explain some conflicting data on the ability of RASFs to undergo FasL- and TRAIL-mediated cell death and suggest that strategies to sensitise RASFs to apoptosis may include the targeting of cell cycle-regulating genes.


Subject(s)
Apoptosis/physiology , Arthritis, Rheumatoid/pathology , Cell Cycle/physiology , Fibroblasts/pathology , Synovial Membrane/pathology , Arthritis, Rheumatoid/metabolism , Cell Proliferation , Fas Ligand Protein/metabolism , Fibroblasts/metabolism , Flow Cytometry , Humans , Synovial Membrane/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism
10.
J Orthop Res ; 25(5): 569-77, 2007 May.
Article in English | MEDLINE | ID: mdl-17262819

ABSTRACT

In osteoarthritis (OA), hepatocyte growth factor (HGF) is supposed to play a role in cartilage repair. Because the development of osteophytes is a major characteristic of OA and thought to be part of an attempted repair process, the purpose of this study was to determine whether HGF may be involved in osteophyte formation. HGF levels in synovial fluids from 41 patients assessed by enzyme immunosorbant assay were correlated with disease severity and osteophyte formation, evaluated by anteroposterior weight-bearing radiographs. Detection of HGF, c-Met, and CD68 in cartilage and synovial tissues was assessed by immunohistochemistry. Effects of HGF on the secretion of TGF-beta1 and BMP-2 by chondrocytes, fibroblast-like synovial cells (FLS), and macrophages as well as HGF-induced secretion of MCP-1 by FLS and chondrocytes were determined by ELISA. HGF was detected in all synovial fluids and concentrations correlated highly with disease severity and osteophyte formation (p < 0.001). Immunohistochemistry revealed weak synovial staining for HGF, whereas increasing numbers of HGF expressing chondrocytes were detected depending on disease severity. In addition, an increased number of macrophages in synovial specimens was observed, which was likewise severity dependent. In a series of subsequent in vitro studies, HGF remarkable induced MCP-1 secretion by FLS in a dose-dependent manner. No effect on TGF-beta1 and BMP-2 secretion by FLS and chondrocytes was evident upon HGF stimulation, whereas secretion of these growth factors by PMA-differentiated THP-1 cells was significantly increased by HGF. The results indicate that HGF may facilitate osteophyte development by promoting MCP-1-mediated entry of monocytes/macrophages into the OA-affected joint and/or by stimulating macrophage-derived growth factors.


Subject(s)
Chemokine CCL2/metabolism , Chondrocytes/physiology , Hepatocyte Growth Factor/metabolism , Osteoarthritis/physiopathology , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cartilage/metabolism , Cartilage/pathology , Cells, Cultured , Chondrocytes/pathology , Female , Femur/metabolism , Femur/pathology , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Monocytes/metabolism , Osteoarthritis/diagnostic imaging , Osteoarthritis/pathology , Proto-Oncogene Proteins c-met/metabolism , Radiography , Severity of Illness Index , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology
11.
Planta ; 224(2): 380-93, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16435132

ABSTRACT

Arabidopsis thaliana L. (Heynh.) plants were grown in low light (150 micromol photons m(-2) s(-1) and 20 degrees C) either in short days (7.5 h photoperiod) or long days (16 h photoperiod), and then transferred into high light and low temperature (350-800 micromol photons m(-2) s(-1) at 12 degrees C). Plants grown in short days responded with a rapid increase in NADP-malate dehydrogenase (EC 1.1.1.82) activation state. However, persisting overreduction revealed a new level of regulation of the malate valve. Activity measurements and Northern-blot analyses indicated that NADP-malate dehydrogenase transcript and protein levels increased within a few hours. Using macroarrays, additional changes in gene expression were identified. Transcript levels for several enzymes of glutathione metabolism and of some photosynthetic genes increased. The cellular glutathione level increased, but its redox state remained unchanged. A different situation was observed in plants grown in long-day conditions. Neither NADP-malate dehydrogenase nor glutathione content changed, but the expression of several antioxidative enzymes increased strongly. We conclude that the endogenous systems that measure day length interact with redox regulation, and override the interpretation of the signals, i.e. they redirect redox-mediated acclimation signals to allow for more efficient light usage and redox poising in short days to systems for the prevention of oxidative damages when grown under long-day conditions.


Subject(s)
Arabidopsis/physiology , Arabidopsis/radiation effects , Light , Photoperiod , Antioxidants/metabolism , Chlorophyll/metabolism , Fluorescence , Gene Expression Profiling , Gene Expression Regulation, Plant , Genes, Plant/genetics , Malate Dehydrogenase (NADP+)/metabolism , Malates/metabolism , Oxidation-Reduction/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...