Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 79(3): 184, 2022 Mar 12.
Article in English | MEDLINE | ID: mdl-35279748

ABSTRACT

The pathogenesis of acute kidney injury (AKI) is associated with the activation of multiple signaling pathways, including Wnt/ß-catenin signaling. However, the mechanism of Wnt/ß-catenin pathway activation in renal interstitial fibroblasts during AKI is unclear. S100 calcium-binding protein A16 (S100A16), a new member of calcium-binding protein S100 family, is a multi-functional signaling factor involved in various pathogenies, including tumors, glycolipid metabolism disorder, and chronic kidney disease (CKD). We investigated the potential participation of S100A16 in Wnt/ß-catenin pathway activation during AKI by subjecting wild-type (WT) and S100A16 knockout (S100A16+/-) mice to the ischemia-reperfusion injury (IRI), and revealed S100A16 upregulation in this model, in which knockout of S100A16 impeded the Wnt/ß-catenin signaling pathway activation and recovered the expression of downstream hepatocyte growth factor (HGF). We also found that S100A16 was highly expressed in Platelet-derived growth factor receptor beta (PDGFRß) positive renal fibroblasts in vivo. Consistently, in rat renal interstitial fibroblasts (NRK-49F cells), both hypoxia/reoxygenation and S100A16 overexpression exacerbated fibroblasts apoptosis and inhibited HGF secretion; whereas S100A16 knockdown or Wnt/ß-catenin pathway inhibitor ICG-001 reversed these changes. Mechanistically, we showed that S100A16 promoted Wnt/ß-catenin signaling activation via the ubiquitylation and degradation of ß-catenin complex members, glycogen synthase kinase 3ß (GSK3ß) and casein kinase 1α (CK1α), mediated by E3 ubiquitin ligase, the HMG-CoA reductase degradation protein 1 (HRD1). Our study identified the S100A16 as a key regulator in the activation of Wnt/ß-catenin signaling pathway in AKI.


Subject(s)
Acute Kidney Injury/pathology , Casein Kinase Ialpha/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , S100 Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Acute Kidney Injury/metabolism , Animals , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Knockout , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , S100 Proteins/antagonists & inhibitors , S100 Proteins/deficiency , S100 Proteins/genetics , Ubiquitination , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , bcl-2-Associated X Protein/metabolism
2.
Int J Med Sci ; 17(14): 2087-2094, 2020.
Article in English | MEDLINE | ID: mdl-32922169

ABSTRACT

Interleukin (IL)-13 plays a key role in the pathogenesis of atopic dermatitis (AD). Our preliminary study demonstrated that forced expression of miR-143 could block IL-13-induced down-regulation of epidermal barrier related proteins in epidermal keratinocytes. As previous studies suggested that miR-143 expression was regulated by mammalian target of rapamycin (mTOR) signaling pathway, we investigated the mechanism of mTOR signaling pathway in the epidermal barrier dysfunction of AD. The HaCaT cells were stimulated by IL-13 and subsequently treated with rapamycin. The expression levels of miR-143, IL-13 receptor α1 (IL-13Rα1), p-mTOR, p-S6K1, p-Akt, and epidermal barrier related proteins were analyzed through RT-qPCR and/or western blotting. The current study showed that IL-13 increased the expression levels of p-mTOR, p-S6K1, and p-Akt, and that rapamycin blocked IL-13-induced down-regulation of miR-143, suppressed the IL-13Rα1 expression and up-regulated the expressions of filaggrin, loricrin, and involucrin in HaCaT cells. This study proposed that IL-13 could activate the mTOR signaling pathway, and confirmed the vital role of mTOR-miR-143 signaling axis in the pathogenesis of AD. It provided solid evidences regarding rapamycin as a potential effective therapeutic option in the management of AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Interleukin-13/metabolism , MicroRNAs/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Filaggrin Proteins , HaCaT Cells , Humans , Interleukin-13/immunology , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/metabolism , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Protein Precursors/deficiency , Protein Precursors/genetics , S100 Proteins/deficiency , S100 Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Sirolimus/therapeutic use , Skin/drug effects , Skin/immunology , Skin/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Up-Regulation/drug effects , Up-Regulation/immunology
4.
Cell Mol Immunol ; 17(10): 1053-1062, 2020 10.
Article in English | MEDLINE | ID: mdl-31467414

ABSTRACT

Toll-like receptors (TLRs) are key pattern recognition receptors that mediate innate immune responses to infection. However, uncontrolled TLR activation can lead to severe inflammatory disorders such as septic shock. The molecular mechanisms through which TLR responses are regulated are not fully understood. Here, we demonstrate an essential function of S100A10 in TLR signaling. S100A10 was constitutively expressed in macrophages, but was significantly downregulated upon TLR activation. S100A10-deficient macrophages were hyperresponsive to TLR stimulation, and S100A10-deficient mice were more sensitive to endotoxin-induced lethal shock and Escherichia coli-induced abdominal sepsis. Mechanistically, S100A10 regulated macrophage inflammatory responses by interfering with the appropriate recruitment and activation of the receptor-proximal signaling components and eventually inhibited TLR-triggered downstream signaling. These findings expand our understanding of TLR signaling and establish S100A10 as an essential negative regulator of TLR function and a potential therapeutic target for treating inflammatory diseases.


Subject(s)
Annexin A2/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Immunity , S100 Proteins/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Annexin A2/deficiency , Cytokines/biosynthesis , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Humans , Inflammation/pathology , Inflammation Mediators/metabolism , Lipopolysaccharides , Macrophages/metabolism , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Phagocytosis/drug effects , Protein Binding , Protein Domains , S100 Proteins/deficiency , Sepsis/pathology , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/metabolism
5.
FASEB J ; 33(1): 455-468, 2019 01.
Article in English | MEDLINE | ID: mdl-30001168

ABSTRACT

External acidity induces catecholamine secretion by inhibiting TASK1-like channels in rat adrenal medullary (AM) cells. TASK channels can function as a heteromer or homomer in the TASK subfamily. In this study, we elucidate the molecular identity of TASK1-like channels in mouse AM cells using gene knockout. Genetic deletion of TASK1, but not TASK3, abolished the depolarizing inward current and catecholamine secretion in response to acidity, whereas it did not affect the resting current level. Immunocytochemistry revealed that AM cells exhibited predominantly TASK1-like and little TASK3-like immunoreactivity. A proximity ligation assay showed that TASK1/3 heteromeric channels were not formed in AM cells or PC12 cells. However, the exogenous expression of p11 in PC12 cells resulted in the heteromeric formation of TASK isoforms, which were mainly located in the cytoplasm, and p11 was not expressed in rat adrenal medullae or PC12 cells. In AM cells, genetic deletion of TASK1 resulted in enhancement of the immunoreactivity of the TALK2 channel, but not TASK3. The results indicate that TASK1 homomeric channels function as acidity sensors in AM cells, and that function is facilitated by the lack of p11 expression.-Inoue, M., Matsuoka, H., Lesage, F., Harada, K. Lack of p11 expression facilitates acidity-sensing function of TASK1 channels in mouse adrenal medullary cells.


Subject(s)
Acid Sensing Ion Channels/physiology , Acids/chemistry , Adrenal Medulla/physiology , Annexin A2/deficiency , Nerve Tissue Proteins/physiology , Potassium Channels, Tandem Pore Domain/physiology , Potassium Channels/physiology , S100 Proteins/deficiency , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PC12 Cells , Rats
6.
Physiol Rep ; 6(15): e13822, 2018 08.
Article in English | MEDLINE | ID: mdl-30101473

ABSTRACT

Calmodulin (CaM) and S100A1 fine-tune skeletal muscle Ca2+ release via opposite modulation of the ryanodine receptor type 1 (RyR1). Binding to and modulation of RyR1 by CaM and S100A1 occurs predominantly at the region ranging from amino acid residue 3614-3640 of RyR1 (here referred to as CaMBD2). Using synthetic peptides, it has been shown that CaM binds to two additional regions within the RyR1, specifically residues 1975-1999 and 4295-4325 (CaMBD1 and CaMBD3, respectively). Because S100A1 typically binds to similar motifs as CaM, we hypothesized that S100A1 could also bind to CaMBD1 and CaMBD3. Our goals were: (1) to establish whether S100A1 binds to synthetic peptides containing CaMBD1 and CaMBD3 using isothermal calorimetry (ITC), and (2) to identify whether S100A1 and CaM modulate RyR1 Ca2+ release activation via sites other than CaMBD2 in RyR1 in its native cellular context. We developed the mouse model (RyR1D-S100A1KO), which expresses point mutation RyR1-L3625D (RyR1D) that disrupts the modulation of RyR1 by CaM and S100A1 at CaMBD2 and also lacks S100A1 (S100A1KO). ITC assays revealed that S100A1 binds with different affinities to CaMBD1 and CaMBD3. Using high-speed Ca2+ imaging and a model for Ca2+ binding and transport, we show that the RyR1D-S100A1KO muscle fibers exhibit a modest but significant increase in myoplasmic Ca2+ transients and enhanced Ca2+ release flux following field stimulation when compared to fibers from RyR1D mice, which were used as controls to eliminate any effect of binding at CaMBD2, but with preserved S100A1 expression. Our results suggest that S100A1, similar to CaM, binds to CaMBD1 and CaMBD3 within the RyR1, but that CaMBD2 appears to be the primary site of RyR1 regulation by CaM and S100A1.


Subject(s)
Calmodulin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , S100 Proteins/physiology , Action Potentials/physiology , Animals , Calcium/metabolism , Calorimetry/methods , Excitation Contraction Coupling/physiology , Male , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , S100 Proteins/deficiency
7.
Diagn Cytopathol ; 44(4): 317-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26799943

ABSTRACT

Intranodal palisaded myofibroblastoma (IPM) is a benign entity, characterized by intranodal proliferation of cells of myofibroblastic origin. It has five distinct histologic features: compressed remnants of lymphoid tissue at the periphery, spindle cells with nuclear palisading, intralesional hemorrhage, amianthoid fibers, and intracellular and extracellular fuchsinophilic bodies. The spindle cells are SMA and vimentin (IHC) positive and are negative for S 100, and has a low proliferative index. Cytologic diagnosis of this lesion is a diagnostic challenge and has to be differentiated from other stroma rich lesions including schwannoma. The FNA smears were reviewed after histopathology to look for any specific features. This report highlights the variable character of spindle cells, presence of unique "amanthiod fibers" and blood vessel within cell clusters in MGG stained FNA smears, which can be a useful diagnostic clue. This observation is being reported for the first time.


Subject(s)
Biomarkers, Tumor/genetics , Neoplasms, Muscle Tissue/diagnosis , Neurilemmoma/diagnosis , Actins/genetics , Biopsy, Fine-Needle , Cell Proliferation , Diagnosis, Differential , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymph Nodes/surgery , Male , Middle Aged , Neoplasms, Muscle Tissue/genetics , Neoplasms, Muscle Tissue/pathology , Neoplasms, Muscle Tissue/surgery , Neurilemmoma/genetics , Neurilemmoma/pathology , Neurilemmoma/surgery , S100 Proteins/deficiency , S100 Proteins/genetics , Vimentin/genetics
8.
Sci Rep ; 5: 14871, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26445893

ABSTRACT

Thymic epithelial cells (TECs) form a 3-dimentional network supporting thymocyte development and maturation. Besides epithelium and thymocytes, heterogeneous fibroblasts are essential components in maintaining thymic microenvironments. However, thymic fibroblast characteristics, development and function remain to be determined. We herein found that thymic non-hematopoietic CD45(-)FSP1(+) cells represent a unique Fibroblast specific protein 1 (FSP1)(-)fibroblast-derived cell subset. Deletion of these cells in FSP1-TK transgenic mice caused thymus atrophy due to the loss of TECs, especially mature medullary TECs (MHCII(high), CD80(+) and Aire(+)). In a cyclophosphamide-induced thymus injury and regeneration model, lack of non-hematopoietic CD45(-)FSP1(+) fibroblast subpopulation significantly delayed thymus regeneration. In fact, thymic FSP1(+) fibroblasts released more IL-6, FGF7 and FSP1 in the culture medium than their FSP1(-) counterparts. Further experiments showed that the FSP1 protein could directly enhance the proliferation and maturation of TECs in the in vitro culture systems. FSP1 knockout mice had significantly smaller thymus size and less TECs than their control. Collectively, our studies reveal that thymic CD45(-)FSP1(+) cells are a subpopulation of fibroblasts, which is crucial for the maintenance and regeneration of TECs especially medullary TECs through providing IL-6, FGF7 and FSP1.


Subject(s)
Epithelial Cells/cytology , Fibroblasts/cytology , Regeneration/physiology , S100 Proteins/genetics , Thymus Gland/cytology , Animals , Animals, Newborn , Cell Communication/physiology , Cell Differentiation , Cell Proliferation , Cyclophosphamide/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fetus , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Interleukin-6/genetics , Interleukin-6/metabolism , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , S100 Calcium-Binding Protein A4 , S100 Proteins/deficiency , Thymus Gland/drug effects , Thymus Gland/metabolism
9.
Med Sci (Paris) ; 31(5): 546-50, 2015 May.
Article in French | MEDLINE | ID: mdl-26059306

ABSTRACT

The comorbidity of depression and cocaine addiction suggests shared mechanisms and anatomical pathways. Specifically, the limbic structures, such as the nucleus accumbens (NAc), play a crucial role in both disorders. P11 (S100A10) is a promising target for manipulating depression and addiction in mice. We summarized the recent genetic and viral strategies used to determine how the titration of p11 levels within the NAc affects hedonic behavior and cocaine reward learning in mice. In particular, p11 in the ChAT+ cells or DRD1+ MSN of the NAc, controls depressive-like behavior or cocaine reward, respectively. Treatments to counter maladaptation of p11 levels in the NAc could provide novel therapeutic opportunities for depression and cocaine addiction in humans.


Subject(s)
Annexin A2/physiology , Depression/epidemiology , Depressive Disorder/epidemiology , Nerve Tissue Proteins/physiology , Nucleus Accumbens/physiopathology , S100 Proteins/physiology , Substance-Related Disorders/epidemiology , Anhedonia/drug effects , Anhedonia/physiology , Animals , Annexin A2/deficiency , Annexin A2/genetics , Appetitive Behavior/physiology , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Cocaine/pharmacology , Cocaine/toxicity , Comorbidity , Depression/physiopathology , Depression/therapy , Depressive Disorder/physiopathology , Depressive Disorder/therapy , Disease Models, Animal , Genetic Therapy , Genetic Vectors/therapeutic use , Humans , Interneurons/drug effects , Interneurons/physiology , Mice , Mice, Knockout , Molecular Targeted Therapy , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Pathways/drug effects , Neural Pathways/physiology , Neurotransmitter Agents/physiology , Nucleus Accumbens/drug effects , Optogenetics , Pleasure/physiology , Prevalence , Protein Transport/drug effects , RNA Interference , Receptors, Neurotransmitter/drug effects , Receptors, Neurotransmitter/metabolism , Reward , S100 Proteins/deficiency , S100 Proteins/genetics , Substance-Related Disorders/physiopathology , Substance-Related Disorders/therapy
10.
Cardiovasc Res ; 105(1): 8-19, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25395393

ABSTRACT

AIMS: S100A1, a 10-kDa, Ca(2+)-binding protein, is expressed in endothelial cells (ECs) and binds eNOS. Its absence is associated with impaired production of nitric oxide (NO) and mild systemic hypertension. As endothelial dysfunction contributes to clinical and experimental pulmonary hypertension (PH), we investigated the impact of deleting S100A1 in mice, on pulmonary haemodynamics, endothelial function, NO production, associated signalling pathways, and apoptosis. METHODS AND RESULTS: Compared with wild-type (WT), S100A1-knock-out mice (KO) exhibited increased right ventricular (RV) weight/body weight ratio and elevated RV pressure in the absence of altered left ventricular filling pressures, accompanied by increase in wall thickness of muscularized pulmonary arteries and a reduction in microvascular perfusion. In isolated lung preparations, KO revealed reduced basal NO, blunted dose-responsiveness to acetylcholine, and augmented basal and angiotensin (AII)-induced pulmonary vascular resistance (R0) compared with WT. Pre-treatment of KO lungs with S100A1 attenuated the AII-induced increase in pulmonary arterial pressure and R0. S100A1-induced phosphorylation of eNOS, Akt, and ERK1/2 is attenuated in pulmonary EC of KO compared with WT. Basal and TNF-α-induced EC apoptosis is greater in KO vs. WT, and cell survival is enhanced by S100A1 treatment. CONCLUSION: Our data demonstrate that the absence of S100A1 results in PH by disruption of its normal capacity to (i) enhance pulmonary EC function by induction of eNOS activity and NO levels via Akt/ERK1/2 pathways and (ii) promote EC survival. The ability of exogenously administered S100A1 to rescue this phenotype makes it an attractive therapeutic target in the treatment of PH.


Subject(s)
Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Pulmonary Artery/physiopathology , S100 Proteins/deficiency , Animals , Apoptosis/physiology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , HSP90 Heat-Shock Proteins/metabolism , Hypertension, Pulmonary/pathology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , S100 Proteins/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Remodeling
11.
Cell Biochem Biophys ; 71(1): 279-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25120023

ABSTRACT

S100A6 (calcyclin), functions in cell cycle progression and differentiation, has been reported to promote the tumorigenesis and malignancy of many types of cancers. Clear cell renal cell carcinoma (ccRCC) is the most common subtype of RCC, lacking both promising prognostic markers and effective therapeutic targets. In our previous study, we have found the elevated S100A6 in the ccRCC tumor tissues, and the differentially expressed genes determined by microarray analysis were found to be strongly related to tumor metastasis after S100A6 knockdown and overexpression in the ccRCC cell line 786-O. The mRNA expression of S100A6 detected by RT-PCR in 6 cell lines and 174 tumor tissues, including 58 metastatic ccRCC and 116 clinicopathological features paired non-metastatic ccRCC (1:2), indicated S100A6 was elevated in the metastatic cells and tumor tissues. The protein expression was consistent with mRNA expression. The biological function of S100A6 in promoting metastasis was determined through overexpression and knockdown of S100A6 in the ccRCC cell lines 786-O, caki-1, and ACHN. In the scratch wound migration assay as well as migration and invasion assays, S100A6 knockdown significantly suppressed the migratory and invasive abilities of tumor cells, whereas overexpression enhanced the malignancy. Further research with the follow-up data of 129 ccRCC patients were analyzed by the Cox regression and survival analysis. The expression of S100A6 was up-regulated in metastatic ccRCC cells. In the metastatic tumor tissues, the expression of S100A6 was also higher than in the non-metastatic tissues. High S100A6 expression might be crucial to promote metastasis in ccRCC by enhancing the ability of tumor cells migration and invasion. In addition, the quantitative mRNA expression of S100A6 in the tumor tissues was an independent risk factor and might be used as a prognostic marker for the metastatic risk of the localized T1-T2 stage ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Cycle Proteins/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , S100 Proteins/metabolism , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk , S100 Calcium Binding Protein A6 , S100 Proteins/deficiency , S100 Proteins/genetics
12.
Eur Rev Med Pharmacol Sci ; 18(22): 3484-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25491625

ABSTRACT

OBJECTIVE: S100A4 and Slug are known to be closely involved in resistance to chemotherapy. Furthermore, Slug signal was regulated by S100A4. Targeted therapy reducing S100A4 expression and Slug pathway activity may overcome the chemoresistance of human cancers. We hypothesized that over-expression of S100A4 and Slug was associated with the resistance to cisplatin of laryngeal carcinoma Hep-2 cells. We explored whether S100A4 silencing inhibited Slug, resulting in sensitization of laryngeal carcinoma Hep-2 cells to cisplatin. MATERIALS AND METHODS: We investigated the effects of S100A4 and Slug silencing by siRNA transfection on chemosensitivity to cisplatin (DDP) in Hep-2 cells in vitro. In order to confirm the correlation between S100A4 and Slug signals, siRNA transfected Hep-2 cells were over-expressed by pSlug transfection, then explored the effect of S100A4 silencing on chemosensitivity to cisplatin (DDP) in Hep-2 cells in vitro. Real-time RT-PCR and Western blotting confirmed the presence of S100A4 mRNA, Slug mRNA and proteins in Hep-2 cells. RESULTS: We found that resistance or insensitivity of Hep-2 cells to cisplatin might be associated with S100A4 and Slug expression. Knockdown of S100A4 and Slug markedly enhanced the cisplatin-induced suppression of Hep-2 cell growth and increased apoptosis. Knockdown of S100A4 may significantly reduce the levels of S100A4 mRNA, Slug mRNA and proteins, in cisplatin-treated Hep-2 cells. Re-expression of Slug in S100A4 siRNA transfected Hep-2 cells restored the cisplatin resistance in the Hep-2 cells. CONCLUSIONS: Overexpression of S100A4 may be associated with the resistance to cisplatin of laryngeal carcinoma Hep-2 cells. Knockdown of S100A4 enhances the sensitivity to cisplatin of laryngeal carcinoma cells via inhibition of Slug expression.


Subject(s)
Laryngeal Neoplasms/metabolism , S100 Proteins/biosynthesis , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques/methods , Humans , Laryngeal Neoplasms/pathology , RNA, Small Interfering/genetics , S100 Calcium-Binding Protein A4 , S100 Proteins/deficiency , S100 Proteins/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
13.
Sci Transl Med ; 6(218): 218ra4, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24401939

ABSTRACT

The identification of diagnostic markers and therapeutic candidate genes in common diseases is complicated by the involvement of thousands of genes. We hypothesized that genes co-regulated with a key gene in allergy, IL13, would form a module that could help to identify candidate genes. We identified a T helper 2 (TH2) cell module by small interfering RNA-mediated knockdown of 25 putative IL13-regulating transcription factors followed by expression profiling. The module contained candidate genes whose diagnostic potential was supported by clinical studies. Functional studies of human TH2 cells as well as mouse models of allergy showed that deletion of one of the genes, S100A4, resulted in decreased signs of allergy including TH2 cell activation, humoral immunity, and infiltration of effector cells. Specifically, dendritic cells required S100A4 for activating T cells. Treatment with an anti-S100A4 antibody resulted in decreased signs of allergy in the mouse model as well as in allergen-challenged T cells from allergic patients. This strategy, which may be generally applicable to complex diseases, identified and validated an important diagnostic and therapeutic candidate gene in allergy.


Subject(s)
Genetic Association Studies , Hypersensitivity/genetics , Hypersensitivity/immunology , S100 Proteins/genetics , Translational Research, Biomedical , Adult , Animals , Antibodies, Blocking/pharmacology , Cell Polarity , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dermatitis/diagnosis , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/prevention & control , Disease Models, Animal , Epitopes/drug effects , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypersensitivity/diagnosis , Hypersensitivity/prevention & control , Immunologic Memory/drug effects , Interleukin-13/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Models, Immunological , Rhinitis, Allergic, Seasonal/diagnosis , Rhinitis, Allergic, Seasonal/genetics , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/pathology , S100 Calcium-Binding Protein A4 , S100 Proteins/deficiency , Th2 Cells/cytology , Th2 Cells/immunology
14.
Am J Physiol Heart Circ Physiol ; 305(9): H1363-72, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23997102

ABSTRACT

Understanding the role of fibroblasts in pathologic conditions is hampered by the absence of specific markers. Fibroblast-specific protein (FSP)1 has been suggested as a fibroblast-specific marker in normal and fibrotic tissues; FSP1 reporter mice and FSP1-Cre-driven gene deletion are considered reliable strategies to investigate fibroblast biology. Because fibroblasts are abundant in normal and injured mammalian hearts, we studied the identity of FSP1(+) cells in the infarcted and remodeling myocardium using mice with green fluorescent protein (GFP) expression driven by the FSP1 promoter. Neonatal and adult mouse hearts had low numbers of FSP1(+) cells. Myocardial infarction induced marked infiltration with FSP1-expressing cells that peaked after 72 h of reperfusion. Using flow cytometry, we identified 50% of FSP1(+) cells as hematopoietic cells; many endothelial cells were also FSP1(+). Increased infiltration with FSP1(+) cells was also noted in the pressure-overloaded myocardium. Although some FSP1(+) cells had fibroblast morphology, >30% were identified as hematopoietic cells, endothelial cells, or vascular smooth muscle cells. In contrast, periostin did not stain leukocytes or vascular cells but labeled spindle-shaped interstitial cells and, as a typical matricellular protein, was deposited in the matrix. CD11b(+) myeloid cells sorted from the infarcted heart had higher FSP1 expression than corresponding CD11b-negative cells, highlighting the predominant expression by hematopoietic cells. FSP1 is not a specific marker for fibroblasts in cardiac remodeling and fibrosis.


Subject(s)
Fibroblasts/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , S100 Proteins/metabolism , Ventricular Remodeling , Animals , Biomarkers/metabolism , CD11b Antigen/metabolism , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Promoter Regions, Genetic , S100 Calcium-Binding Protein A4 , S100 Proteins/deficiency , S100 Proteins/genetics
15.
Biochim Biophys Acta ; 1833(12): 2653-2663, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23830916

ABSTRACT

OBJECTIVE: S100A4 is a Ca-binding protein that regulates cell growth, survival, and motility. The abundant expression of S100A4 in rheumatiod arthritis contributes to the invasive growth of joint tissue and to bone damage. In the present study, we analysed the role of S100A4 in bone homeostasis. METHODS: Peripheral quantitative computed tomography and histomorphometric analysis were performed in mice lacking the entire S100A4 protein (S100A4KO) and in wild-type (WT) counterparts treated with shRNA-lentiviral constructs targeting S100A4 (S100A4-shRNA). Control groups consisted of sex-matched WT counterparts and WT mice treated with a non-targeting RNA construct. RESULTS: S100A4 deficiency was associated with higher trabecular and cortical bone mass, increased number and thickness of trabeculi combined with larger periosteal circumference and higher predicted bone strength. S100A4 inhibition by shRNA led to an increase in cortical bone in WT mice. S100A4-deficieny was associated with a reduced number of functional osteoclasts. S100A4KO and S100A4-shRNA-treated bone marrow progenitors gave rise to a large number of small TRAP+ cells with few nuclei and few pseudopodial processes. Poor osteoclastogenesis and the low resorptive capacity in S100A4Ko mice may be linked to low levels of surface integrins, impaired adhesion capacity, and poor multinucleation in S100A4-deficient osteoclasts, as well as a low content of proteolytic enzymes cathepsin K and MMP3 and MMP9 to break down the organic matrix. CONCLUSION: S100A4 emerges as a negative regulator of bone metabolism potentially responsible for the excessive bone turnover in conditions marked by high levels of S100A4 protein, such as inflammation and rheumatoid arthritis.


Subject(s)
Bone Resorption/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , S100 Proteins/metabolism , Animals , Bone Remodeling , Bone Resorption/complications , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Membrane/metabolism , Cell Shape , Extracellular Matrix/metabolism , Integrins/metabolism , Matrix Metalloproteinases/metabolism , Mice , Organ Size , Osteolysis/complications , Osteolysis/pathology , Osteolysis/physiopathology , Phenotype , S100 Calcium-Binding Protein A4 , S100 Proteins/deficiency
16.
PLoS One ; 8(1): e55122, 2013.
Article in English | MEDLINE | ID: mdl-23383075

ABSTRACT

BACKGROUND: S100A4, a member of the calcium-binding proteins, is dramatically elevated in a variety of fibrotic diseases. Areca quid chewing is the most important etiological factor in the pathogenesis of oral submucous fibrosis (OSF). OSF has been considered as a pre-cancerous condition of oral mucosa. The aim of this study was to determine the critical role of S100A4 expression in the pathogenesis of OSF both in vitro and in vivo. METHODOLOGY/PRINCIPAL FINDING: Thirty OSF tissues from areca quid chewers and ten normal buccal mucosa samples without areca quid chewing were analyzed by using immunohistochemistry for S100A4 expression in vivo. Collagen gel contraction capability and expression of tissue inhibitor of metalloproteinases 1 (TIMP1)/MMP9 in arecoline-stimulated BMFs with S100A4 knockdown was presented in vitro. Initially, S100A4 expression was higher in areca quid chewing-associated OSF specimens than normal buccal mucosa specimens (p = 0.001). Arecoline, a major areca nut alkaloid, led to dose- and time-dependent elevation of S100A4 expression in normal buccal mucosa fibroblasts BMFs (p<0.05). The additions of pharmacological agents rapamycin (mTOR inhibitor), PD98059 (ERK inhibitor), and Bay117082 (NF-κB inhibitor) were found to inhibit arecoline-induced S100A4 expression (p<0.05) in BMFs. Down-regulation of S100A4 by lentiviral infection significantly reversed arecoline-induced collagen gel contraction and TIMP1/MMP9 expression. CONCLUSION/SIGNIFICANCE: These results suggest that S100A4 expression is significantly up-regulated in OSF specimens. Arecoline-induced S100A4 expression was down-regulated by rapamycin, PD98059, and Bay117082. Targeting S100A4 might be a potential therapeutic target for OSF through TIMP1/MMP9 down-regulation.


Subject(s)
Arecoline/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Mouth Mucosa/cytology , Oral Submucous Fibrosis/pathology , S100 Proteins/genetics , Up-Regulation/drug effects , Collagen/metabolism , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/pathology , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 9/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mouth Mucosa/pathology , NF-kappa B/metabolism , Oral Submucous Fibrosis/chemically induced , Oral Submucous Fibrosis/genetics , S100 Calcium-Binding Protein A4 , S100 Proteins/deficiency , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors , Tissue Inhibitor of Metalloproteinase-1/genetics
18.
Circ Res ; 112(1): 66-78, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23048072

ABSTRACT

RATIONALE: Mice lacking the EF-hand Ca2+ sensor S100A1 display endothelial dysfunction because of distorted Ca2+ -activated nitric oxide (NO) generation. OBJECTIVE: To determine the pathophysiological role of S100A1 in endothelial cell (EC) function in experimental ischemic revascularization. METHODS AND RESULTS: Patients with chronic critical limb ischemia showed almost complete loss of S100A1 expression in hypoxic tissue. Ensuing studies in S100A1 knockout (SKO) mice subjected to femoral artery resection unveiled insufficient perfusion recovery and high rates of autoamputation. Defective in vivo angiogenesis prompted cellular studies in SKO ECs and human ECs, with small interfering RNA-mediated S100A1 knockdown demonstrating impaired in vitro and in vivo proangiogenic properties (proliferation, migration, tube formation) and attenuated vascular endothelial growth factor (VEGF)-stimulated and hypoxia-stimulated endothelial NO synthase (eNOS) activity. Mechanistically, S100A1 deficiency compromised eNOS activity in ECs by interrupted stimulatory S100A1/eNOS interaction and protein kinase C hyperactivation that resulted in inhibitory eNOS phosphorylation and enhanced VEGF receptor-2 degradation with attenuated VEGF signaling. Ischemic SKO tissue recapitulated the same molecular abnormalities with insufficient in vivo NO generation. Unresolved ischemia entailed excessive VEGF accumulation in SKO mice with aggravated VEGF receptor-2 degradation and blunted in vivo signaling through the proangiogenic phosphoinositide-3-kinase/Akt/eNOS cascade. The NO supplementation strategies rescued defective angiogenesis and salvaged limbs in SKO mice after femoral artery resection. CONCLUSIONS: Our study shows for the first time downregulation of S100A1 expression in patients with critical limb ischemia and identifies S100A1 as critical for EC function in postnatal ischemic angiogenesis. These findings link its pathological plasticity in critical limb ischemia to impaired neovascularization, prompting further studies to probe the microvascular therapeutic potential of S100A1.


Subject(s)
Endothelial Cells/enzymology , Ischemia/enzymology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , S100 Proteins/deficiency , Aged , Aged, 80 and over , Animals , Calcium/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Female , Hindlimb , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Ischemia/drug therapy , Ischemia/genetics , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Skeletal/pathology , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Regional Blood Flow , S100 Proteins/genetics , Signal Transduction , Time Factors , Transfection , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Mol Psychiatry ; 18(10): 1096-105, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23032875

ABSTRACT

Cognitive impairments are common in depression and involve dysfunctional serotonin neurotransmission. The 5-HT1B receptor (5-HT(1B)R) regulates serotonin transmission, via presynaptic receptors, but can also affect transmitter release at heterosynaptic sites. This study aimed at investigating the roles of the 5-HT(1B)R, and its adapter protein p11, in emotional memory and object recognition memory processes by the use of p11 knockout (p11KO) mice, a genetic model for aspects of depression-related states. 5-HT(1B)R agonist treatment induced an impairing effect on emotional memory in wild type (WT) mice. In comparison, p11KO mice displayed reduced long-term emotional memory performance. Unexpectedly, 5-HT(1B)R agonist stimulation enhanced memory in p11KO mice, and this atypical switch was reversed after hippocampal adeno-associated virus mediated gene transfer of p11. Notably, 5-HT(1B)R stimulation increased glutamatergic neurotransmission in the hippocampus in p11KO mice, but not in WT mice, as measured by both pre- and postsynaptic criteria. Magnetic resonance spectroscopy demonstrated global hippocampal reductions of inhibitory GABA, which may contribute to the memory enhancement and potentiation of pre- and post-synaptic measures of glutamate transmission by a 5-HT(1B)R agonist in p11KO mice. It is concluded that the level of hippocampal p11 determines the directionality of 5-HT(1B)R action on emotional memory processing and modulates hippocampal functionality. These results emphasize the importance of using relevant disease models when evaluating the role of serotonin neurotransmission in cognitive deficits related to psychiatric disorders.


Subject(s)
Annexin A2/physiology , Avoidance Learning/physiology , Emotions/physiology , Hippocampus/physiology , Memory/physiology , Receptor, Serotonin, 5-HT1B/physiology , S100 Proteins/physiology , Animals , Annexin A2/deficiency , Annexin A2/genetics , Avoidance Learning/drug effects , Depression/physiopathology , Disease Models, Animal , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Genes, Reporter , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation/drug effects , Presynaptic Terminals/metabolism , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , Reaction Time , Receptors, AMPA/metabolism , Recombinant Fusion Proteins/metabolism , S100 Proteins/deficiency , S100 Proteins/genetics , Serotonin 5-HT1 Receptor Agonists/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Transduction, Genetic
20.
Cancer Res ; 71(21): 6676-83, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22042827

ABSTRACT

Macrophages are critical drivers of tumor growth, invasion, and metastasis. Movement of macrophages into tumors requires the activity of cell surface proteases such as plasmin. In this study, we offer genetic evidence that plasminogen receptor S100A10 is essential for recruitment of macrophages to the tumor site. Growth of murine Lewis lung carcinomas or T241 fibrosarcomas was dramatically reduced in S100A10-deficient mice compared with wild-type mice. The tumor growth deficit corresponded with a decrease in macrophage density that could be rescued by intraperitoneal injection of wild-type but not S100A10-deficient macrophages. Notably, macrophages of either genotype could rescue tumor growth if they were injected into the tumor itself, establishing that S100A10 was required specifically for the migratory capability needed for tumor homing. Conversely, selective depletion of macrophages from wild-type mice phenocopied the tumor growth deficit seen in S100A10-deficient mice. Together, our findings show that S100A10 is essential and sufficient for macrophage migration to tumor sites, and they define a novel rate-limiting step in tumor progression.


Subject(s)
Annexin A2/physiology , Carcinoma, Lewis Lung/pathology , Fibrosarcoma/pathology , Macrophages/physiology , Neoplasm Proteins/physiology , S100 Proteins/physiology , Animals , Annexin A2/deficiency , Annexin A2/genetics , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cell Movement/physiology , Disease Progression , Enzyme Activation , Female , Fibrosarcoma/blood supply , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Macrophages, Peritoneal/physiology , Macrophages, Peritoneal/transplantation , Mice , Mice, Knockout , Neovascularization, Pathologic/pathology , Phenotype , Plasminogen/metabolism , S100 Proteins/deficiency , S100 Proteins/genetics , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL
...