Ignet
Search (e.g., vaccine, IFNG): Help
About
Home
Introduction
Statistics
Programs
Dignet
Gene
GenePair
BioSummarAI
Help & Docs
Documents
Help
FAQs
Links
Acknowledge
Disclaimer
Contact Us
UM Logo

UMMS Logo

UMMS Logo

Gene Information

Gene symbol: KITLG

Gene name: KIT ligand

HGNC ID: 6343

Synonyms: SCF, SF, Kitl, KL-1, FPH2

Related Genes

# Gene Symbol Number of hits
1 ABCB1 1 hits
2 ABCG2 1 hits
3 ADIPOQ 1 hits
4 AKT1 1 hits
5 C2orf3 1 hits
6 CASP3 1 hits
7 CD34 1 hits
8 CDH15 1 hits
9 CDKN1B 1 hits
10 CRP 1 hits
11 CSF1R 1 hits
12 CSF2 1 hits
13 CSF3 1 hits
14 CUL1 1 hits
15 EPO 1 hits
16 FAS 1 hits
17 FASLG 1 hits
18 FBXO2 1 hits
19 FBXW10 1 hits
20 FBXW7 1 hits
21 FGF2 1 hits
22 FLT3 1 hits
23 HBB 1 hits
24 HBG1 1 hits
25 IGF1 1 hits
26 IL18 1 hits
27 IL3 1 hits
28 IL4 1 hits
29 IL6 1 hits
30 IL7 1 hits
31 IL7R 1 hits
32 INS 1 hits
33 ISL1 1 hits
34 KDR 1 hits
35 KIT 1 hits
36 KRT124P 1 hits
37 LIF 1 hits
38 MAP3K7 1 hits
39 MAPK1 1 hits
40 MAPK3 1 hits
41 NES 1 hits
42 NOS3 1 hits
43 NPPC 1 hits
44 NPR3 1 hits
45 NR0B2 1 hits
46 PARK2 1 hits
47 PIK3CA 1 hits
48 PPARGC1A 1 hits
49 PSMD9 1 hits
50 PTPRC 1 hits
51 RHOD 1 hits
52 RNF123 1 hits
53 RPS27A 1 hits
54 SIRPA 1 hits
55 SKP1 1 hits
56 SKP2 1 hits
57 SOX17 1 hits
58 TAF8 1 hits
59 TGFA 1 hits
60 TGFB1 1 hits
61 THPO 1 hits
62 THY1 1 hits
63 TNF 1 hits
64 TP53 1 hits
65 VEGFA 1 hits
66 VWF 1 hits

Related Sentences

# PMID Sentence
1 9609630 Altered expression of bladder mast cell growth factor receptor (c-kit) in interstitial cystitis.
2 10339480 Groups of sublethally irradiated NOD/SCID mice were injected with either 35,000, 20,000, and 10,000 unmanipulated CD34(+) CB cells, which were cryopreserved at the start of cultures, or the cryopreserved cells expanded from 35,000, 20,000, or 10,000 CD34(+) cells for 4, 8, and 12 weeks in the presence of a combination of early acting recombinant growth factors (flt 3/flk2 ligand [FL] + megakaryocyte growth and development factor [MGDF] +/- stem cell factor [SCF] +/- interleukin-6 [IL-6]).
3 10482993 Recently, we developed a xenogenic coculture system involving HUCB-CD34+ cells and murine bone marrow stromal cells, HESS-5 cells, in combination with human interleukin-3 and stem cell factor.
4 10560908 Southern blotting demonstrated transfer of the neo(r) gene to 30% to 80% of leukemic blasts when cells were cultured for 48 hours in the presence of IL-3 and steel factor (SF) prior to 48-hour coculture with viral producers.
5 10609654 Medium for the ex vivo cultures contained either serum and six recombinant human hematopoietic growth factors (GFs), including Flt-3 ligand (FL), Kit ligand (KL = stem cell factor), thrombopoietin (Tpo), interleukin 3 (IL-3), granulocyte colony-stimulating factor (G-CSF), and interleukin 6 (IL-6), or a serum-free medium containing only FL, KL, and Tpo.
6 11177540 CD34+ cells isolated from human cord blood were transduced by exposure to virus-containing supernatants on fibronectin fragments and in the presence of stem cell factor, interleukin 6, Flt-3 ligand, and thrombopoietin, and then transplanted into nonobese diabetic/SCID mice.
7 11238136 Peripheral blood stem cells from 2 rhesus monkeys were collected, CD34-enriched, split into 2 portions, and transduced with either MDR1 vectors or neo vectors and continued in culture for a total of 10 days before reinfusion.
8 11238136 After 3 cycles of stem cell factor/granulocyte colony-stimulating factor, there were no changes in the levels of MDR1 vector- or neo vector-containing cells.
9 11369635 Umbilical cord blood (UCB) CD34(+) cells were cultured in transwells above AFT024 feeders with fetal-liver-tyrosine-kinase (FL) + stem cell factor (SCF) + interleukin 7 (IL-7), or FL + thrombopoietin (Tpo).
10 11372756 The glucoprotein130 signal, activated by a complex of interleukin 6 (IL-6) and soluble IL-6 receptor (IL-6/sIL-6R), acts dramatically in synergy with the c-Kit or Flk2/Flt3 signal to expand immature human HSCs.
11 11372756 The proportion of human CD45+ cells in recipient marrow was 10 times higher in animals receiving the cultured cells with stem cell factor, Flk2/Flt3 ligand, thrombopoietin, and IL-6/sIL-6R than in those receiving comparable numbers of fresh cord blood CD34+ cells.
12 11522667 Indeed, upon treatment with the KIT ligand (KITL), the extracellular signal-regulated protein kinase was phosphorylated, and the expression of early responsive genes was induced.
13 11994455 In GM-CSF/IL-4-supplemented bone marrow cultures, DC developed in significantly greater numbers from NOD than from NOR, BALB/c, and BL/6 mice.
14 11994455 Likewise, DC developed in greater numbers from sorted (lineage(-)IL-7Ralpha(-)SCA-1(-)c-kit(+)) NOD myeloid progenitors in either GM-CSF/IL-4 or GM-CSF/stem cell factor (SCF)/TNF-alpha. [(3)H]TdR incorporation indicated that the increased generation of NOD DC was due to higher levels of myeloid progenitor proliferation.
15 11994455 Consistent with these findings, NOD and NOR mice had increased numbers of DC in blood and thymus and NOD had an increased proportion of the putative myeloid DC (CD11c(+)CD11b(+)) subset within spleen.
16 12088865 Insulin but not insulin-like growth factor-1 promotes the primordial to primary follicle transition.
17 12088865 The current study utilizes a rat ovarian organ culture system to investigate the role of insulin and insulin-like growth factor-1 (IGF-1) in this process.
18 12088865 Ovaries were also treated with epidermal growth factor (EGF) and hepatocyte growth factor (HGF) and neither had an effect on the primordial to primary follicle transition.
19 12088865 Previous experiments have shown that kit ligand (KL), basic fibroblast growth factor (bFGF) and leukemia inhibitory factor (LIF) promote the primordial to primary follicle transition.
20 12088865 Insulin was shown to have an additive effect with KL and LIF, but not bFGF.
21 12088865 The fact that insulin can influence the primordial to primary follicle transition at low concentrations (i.e. 5 ng/ml) and that IGF-1 has no effect suggests that insulin is acting at the insulin receptor, not the IGF-1 receptor.
22 12088865 The observation that insulin has an additive effect with KL and LIF, but not bFGF, suggests the insulin's site of action is likely the oocyte.
23 12204842 Mean volumes (+/- SD) for IAF, SCF and TAF were 10.5 l (+/- 5.0 l), 15.1 l (+/-7.3 l) and 25.7 l (+/-11.5 l), respectively.
24 12204842 Reliability of measurements was excellent for both observers (observer I: intraobserver reliability IAF, SCF and TAF: r = 0.999, r = 0.999 r = 1.0, observer II: r = 0.999, r = 0.999 and r = 1.0), as well as between both observers (interobserver reliability IAF, SCF, and TAF: r = 0.999, r = 0.999, r = 1.0).
25 12204842 Mean volumes (+/- SD) for IAF, SCF and TAF were 10.5 l (+/- 5.0 l), 15.1 l (+/-7.3 l) and 25.7 l (+/-11.5 l), respectively.
26 12204842 Reliability of measurements was excellent for both observers (observer I: intraobserver reliability IAF, SCF and TAF: r = 0.999, r = 0.999 r = 1.0, observer II: r = 0.999, r = 0.999 and r = 1.0), as well as between both observers (interobserver reliability IAF, SCF, and TAF: r = 0.999, r = 0.999, r = 1.0).
27 12208734 Cellular invasion induced by 10 ng/ml stem cell factor (EC(50) = 3 ng/ml) in HT29 cells was blocked by 1 micro M STI571 (IC(50) = 56 nM) and pharmacological inhibitors of several oncogenic signaling pathways, namely, phosphatidylinositol 3-kinase (LY294002), Rho GTPases (Clostridium botulinum exoenzyme C3 transferase), and Rho-kinase (Y27632).
28 12393703 Fetal hemoglobin modulation during human erythropoiesis: stem cell factor has "late" effects related to the expression pattern of CD117.
29 12393703 The average HbF/hemoglobin A (HbA) ratio was 30.9% +/- 18.7% in cultures containing SCF compared with 4.1% +/- 2.2% in those grown with erythropoietin (EPO) alone (P = 8.5E-8).
30 12393703 To further investigate the hemoglobin-modulating effects of SCF, we examined the surface expression pattern of the SCF receptor, CD117, among maturing erythroblasts.
31 12393703 The rise in CD117 expression to high levels mirrored that of the transferrin receptor (CD71), and the subsequent reduction in CD117 was inversely related to increases in expression of glycophorin A.
32 12393703 Fetal hemoglobin modulation during human erythropoiesis: stem cell factor has "late" effects related to the expression pattern of CD117.
33 12393703 The average HbF/hemoglobin A (HbA) ratio was 30.9% +/- 18.7% in cultures containing SCF compared with 4.1% +/- 2.2% in those grown with erythropoietin (EPO) alone (P = 8.5E-8).
34 12393703 To further investigate the hemoglobin-modulating effects of SCF, we examined the surface expression pattern of the SCF receptor, CD117, among maturing erythroblasts.
35 12393703 The rise in CD117 expression to high levels mirrored that of the transferrin receptor (CD71), and the subsequent reduction in CD117 was inversely related to increases in expression of glycophorin A.
36 12393703 Fetal hemoglobin modulation during human erythropoiesis: stem cell factor has "late" effects related to the expression pattern of CD117.
37 12393703 The average HbF/hemoglobin A (HbA) ratio was 30.9% +/- 18.7% in cultures containing SCF compared with 4.1% +/- 2.2% in those grown with erythropoietin (EPO) alone (P = 8.5E-8).
38 12393703 To further investigate the hemoglobin-modulating effects of SCF, we examined the surface expression pattern of the SCF receptor, CD117, among maturing erythroblasts.
39 12393703 The rise in CD117 expression to high levels mirrored that of the transferrin receptor (CD71), and the subsequent reduction in CD117 was inversely related to increases in expression of glycophorin A.
40 12915194 Mean volumes (+/- SD) for IAF, SCF and TAF were 10.5 1 (+/- 5.0 1), 15.1 1 (+/-7.3 1) and 25.7 1 (+/-11.5 1), respectively.
41 14592835 To better understand the relationship between the expression of this fetal protein and growth, donated human erythroid progenitor cells were cultured in the presence of erythropoietin (EPO) plus the growth-modifying cytokine stem cell factor (SCF), and several growth-related signaling pathways were interrogated.
42 14592835 Only the MEK1/2 inhibitor (PD98059) demonstrated significant effects on fetal hemoglobin.
43 14592835 In the absence of PD98059, levels of fetal hemoglobin averaged 27.4% +/- 7.9% in EPO+SCF compared with 1.26% +/- 1.7% in EPO alone (P =.02).
44 14592835 Western blot analyses revealed that SCF was required for phosphorylation of MEK and p44MAPK in this setting, and quantitative polymerase chain reaction demonstrated a significant increase in gamma-globin mRNA.
45 14592835 To better understand the relationship between the expression of this fetal protein and growth, donated human erythroid progenitor cells were cultured in the presence of erythropoietin (EPO) plus the growth-modifying cytokine stem cell factor (SCF), and several growth-related signaling pathways were interrogated.
46 14592835 Only the MEK1/2 inhibitor (PD98059) demonstrated significant effects on fetal hemoglobin.
47 14592835 In the absence of PD98059, levels of fetal hemoglobin averaged 27.4% +/- 7.9% in EPO+SCF compared with 1.26% +/- 1.7% in EPO alone (P =.02).
48 14592835 Western blot analyses revealed that SCF was required for phosphorylation of MEK and p44MAPK in this setting, and quantitative polymerase chain reaction demonstrated a significant increase in gamma-globin mRNA.
49 14592835 To better understand the relationship between the expression of this fetal protein and growth, donated human erythroid progenitor cells were cultured in the presence of erythropoietin (EPO) plus the growth-modifying cytokine stem cell factor (SCF), and several growth-related signaling pathways were interrogated.
50 14592835 Only the MEK1/2 inhibitor (PD98059) demonstrated significant effects on fetal hemoglobin.
51 14592835 In the absence of PD98059, levels of fetal hemoglobin averaged 27.4% +/- 7.9% in EPO+SCF compared with 1.26% +/- 1.7% in EPO alone (P =.02).
52 14592835 Western blot analyses revealed that SCF was required for phosphorylation of MEK and p44MAPK in this setting, and quantitative polymerase chain reaction demonstrated a significant increase in gamma-globin mRNA.
53 14701835 Ubiquitination-mediated regulation of biosynthesis of the adhesion receptor SHPS-1 in response to endoplasmic reticulum stress.
54 14701835 We now show that SHP substrate-1 (SHPS-1), a transmembrane glycoprotein that regulates cytoskeletal reorganization and cell-cell communication, is a physiological substrate for the Skp1-Cullin1-NFB42-Rbx1 (SCF(NFB42)) E3 ubiquitin ligase, a proposed mediator of ER-associated degradation.
55 14701835 SCF(NFB42) mediated the polyubiquitination of immature SHPS-1 and its degradation by the proteasome.
56 14701835 Ectopic expression of NFB42 both suppressed the formation of aggresome-like structures and the phosphorylation of the translational regulator eIF2alpha induced by overproduction of SHPS-1 as well as increased the amount of mature SHPS-1 at the cell surface.
57 14701835 Our results suggest that SCF(NFB42) regulates SHPS-1 biosynthesis in response to ER stress.
58 14701835 Ubiquitination-mediated regulation of biosynthesis of the adhesion receptor SHPS-1 in response to endoplasmic reticulum stress.
59 14701835 We now show that SHP substrate-1 (SHPS-1), a transmembrane glycoprotein that regulates cytoskeletal reorganization and cell-cell communication, is a physiological substrate for the Skp1-Cullin1-NFB42-Rbx1 (SCF(NFB42)) E3 ubiquitin ligase, a proposed mediator of ER-associated degradation.
60 14701835 SCF(NFB42) mediated the polyubiquitination of immature SHPS-1 and its degradation by the proteasome.
61 14701835 Ectopic expression of NFB42 both suppressed the formation of aggresome-like structures and the phosphorylation of the translational regulator eIF2alpha induced by overproduction of SHPS-1 as well as increased the amount of mature SHPS-1 at the cell surface.
62 14701835 Our results suggest that SCF(NFB42) regulates SHPS-1 biosynthesis in response to ER stress.
63 14701835 Ubiquitination-mediated regulation of biosynthesis of the adhesion receptor SHPS-1 in response to endoplasmic reticulum stress.
64 14701835 We now show that SHP substrate-1 (SHPS-1), a transmembrane glycoprotein that regulates cytoskeletal reorganization and cell-cell communication, is a physiological substrate for the Skp1-Cullin1-NFB42-Rbx1 (SCF(NFB42)) E3 ubiquitin ligase, a proposed mediator of ER-associated degradation.
65 14701835 SCF(NFB42) mediated the polyubiquitination of immature SHPS-1 and its degradation by the proteasome.
66 14701835 Ectopic expression of NFB42 both suppressed the formation of aggresome-like structures and the phosphorylation of the translational regulator eIF2alpha induced by overproduction of SHPS-1 as well as increased the amount of mature SHPS-1 at the cell surface.
67 14701835 Our results suggest that SCF(NFB42) regulates SHPS-1 biosynthesis in response to ER stress.
68 15367428 Despite their opposite effects on growth, stem cell factor (SCF) and transforming growth factor beta (TGF-B) had synergistic effects with respect to fetal hemoglobin (HbF): average HbF/HbF + adult hemoglobin (HbA) ratio in erythropoietin (EPO) = 1.4 +/- 1.0%; EPO + TGF-B = 10.8 +/- 1.9%; EPO + SCF = 19.1 +/- 6.2%; and EPO + SCF + TGF-B (EST) = 39.3 +/- 6.3%.
69 15367428 Polymerase chain reaction (PCR) revealed significant increases in gamma-globin transcripts that were balanced by reduced beta-globin transcripts.
70 16305063 Cells expressing the stem cell factor receptor, c-kit, contribute to neoangiogenesis in diabetes.
71 16305063 Origins from bone marrow and endothelial cell differentiation were identified by immunolabelling for the stem cell factor receptor, c-kit and von Willebrand factor (vWF), respectively.
72 16305063 Many of these c-kit+ cells also showed positive immunolabelling for vWF, consistent with endothelial differentiation.
73 16305063 Cells expressing the stem cell factor receptor, c-kit, contribute to neoangiogenesis in diabetes.
74 16305063 Origins from bone marrow and endothelial cell differentiation were identified by immunolabelling for the stem cell factor receptor, c-kit and von Willebrand factor (vWF), respectively.
75 16305063 Many of these c-kit+ cells also showed positive immunolabelling for vWF, consistent with endothelial differentiation.
76 16460677 Human adipose tissue-derived mesenchymal stem cells differentiate into insulin, somatostatin, and glucagon expressing cells.
77 16460677 During the proliferation period, the cells expressed the stem cell markers nestin, ABCG2, SCF, Thy-1 as well as the pancreatic endocrine transcription factor Isl-1.
78 16460677 Using quantitative PCR a down-regulation of ABCG2 and up-regulation of pancreatic developmental transcription factors Isl-1, Ipf-1, and Ngn3 were observed together with induction of the islet hormones insulin, glucagon, and somatostatin.
79 16781008 Regulated protein destruction involving SCF (Skp1/Cullin/F-box, E3 ubiquitin ligase) complexes is required for multicellular development of Dictyostelium discoideum.
80 16781008 Our data also indicate that CSN5 (and CSN2) are essential for cell proliferation in Dictyostelium, a phenotype similar to that of multicellular organisms, but distinct from that of the yeasts.
81 17082193 Skp2 controls adipocyte proliferation during the development of obesity.
82 17082193 The latter effect was found to be accompanied by up-regulation of expression of the gene for the F-box protein Skp2 as well as by downregulation of the cyclin-dependent kinase inhibitor p27(Kip1), a principal target of the SCF(Skp2) ubiquitin ligase, in white adipose tissue.
83 17082193 Ablation of Skp2 protected mice from the development of obesity induced either by a high fat diet or by the lethal yellow agouti (A(y)) mutation, and this protective action was due to inhibition of the increase in adipocyte number without an effect on adipocyte hypertrophy.
84 17082193 The reduction in the number of adipocyte caused by Skp2 ablation also inhibited the development of obesity-related insulin resistance in the A(y) mutant mice, although the reduced number of beta cells and reduced level of insulin secretion in Skp2-deficient mice resulted in glucose intolerance.
85 17082193 Our observations thus indicate that Skp2 controls adipocyte proliferation during the development of obesity.
86 17585379 The number of cells expressing the receptor for Stem Cell Factor (c-kit) and their rate of proliferation were preserved in the left ventricle while the atrial storage of these primitive cells was severely reduced by diabetes-induced oxidative stress.
87 17585379 Despite a down-regulation of Connexin43 and over-expression of both Connexin40 and Connexin45, the junctional proteins were normally distributed in diabetic ventricular myocardium,justifying the preserved tissue excitability and conduction velocity.
88 18198341 SCFCdc4 acts antagonistically to the PGC-1alpha transcriptional coactivator by targeting it for ubiquitin-mediated proteolysis.
89 18198341 Peroxisome proliferator-activated receptor gamma (PPARgamma) coactivator-1alpha (PGC-1alpha) is a highly regulated transcriptional coactivator that coordinates energy metabolism in mammals.
90 18198341 We identified SCF(Cdc4) as an E3 ubiquitin ligase that regulates PGC-1alpha through ubiquitin-mediated proteolysis.
91 18198341 PGC-1alpha contains two Cdc4 phosphodegrons that bind Cdc4 when phosphorylated by Glycogen Synthase Kinase 3beta (GSK3beta) and p38 MAPK, leading to SCF(Cdc4)-dependent ubiquitylation and proteasomal degradation of PGC-1alpha.
92 18198341 Furthermore, SCF(Cdc4) negatively regulates PGC-1alpha-dependent transcription.
93 18198341 These results suggest that attenuation of SCF(Cdc4)-dependent proteasomal degradation of PGC-1alpha has a role in mediating the PGC-1alpha-dependent transcriptional response to oxidative stress.
94 18198341 SCFCdc4 acts antagonistically to the PGC-1alpha transcriptional coactivator by targeting it for ubiquitin-mediated proteolysis.
95 18198341 Peroxisome proliferator-activated receptor gamma (PPARgamma) coactivator-1alpha (PGC-1alpha) is a highly regulated transcriptional coactivator that coordinates energy metabolism in mammals.
96 18198341 We identified SCF(Cdc4) as an E3 ubiquitin ligase that regulates PGC-1alpha through ubiquitin-mediated proteolysis.
97 18198341 PGC-1alpha contains two Cdc4 phosphodegrons that bind Cdc4 when phosphorylated by Glycogen Synthase Kinase 3beta (GSK3beta) and p38 MAPK, leading to SCF(Cdc4)-dependent ubiquitylation and proteasomal degradation of PGC-1alpha.
98 18198341 Furthermore, SCF(Cdc4) negatively regulates PGC-1alpha-dependent transcription.
99 18198341 These results suggest that attenuation of SCF(Cdc4)-dependent proteasomal degradation of PGC-1alpha has a role in mediating the PGC-1alpha-dependent transcriptional response to oxidative stress.
100 18198341 SCFCdc4 acts antagonistically to the PGC-1alpha transcriptional coactivator by targeting it for ubiquitin-mediated proteolysis.
101 18198341 Peroxisome proliferator-activated receptor gamma (PPARgamma) coactivator-1alpha (PGC-1alpha) is a highly regulated transcriptional coactivator that coordinates energy metabolism in mammals.
102 18198341 We identified SCF(Cdc4) as an E3 ubiquitin ligase that regulates PGC-1alpha through ubiquitin-mediated proteolysis.
103 18198341 PGC-1alpha contains two Cdc4 phosphodegrons that bind Cdc4 when phosphorylated by Glycogen Synthase Kinase 3beta (GSK3beta) and p38 MAPK, leading to SCF(Cdc4)-dependent ubiquitylation and proteasomal degradation of PGC-1alpha.
104 18198341 Furthermore, SCF(Cdc4) negatively regulates PGC-1alpha-dependent transcription.
105 18198341 These results suggest that attenuation of SCF(Cdc4)-dependent proteasomal degradation of PGC-1alpha has a role in mediating the PGC-1alpha-dependent transcriptional response to oxidative stress.
106 18772608 Mechano growth factor (MGF) is an alternatively spliced variant of insulin-like growth factor-I (IGF-I).
107 18772608 In the present study, we examined the effect of growth hormone (GH) on MGF expression in C2C12 mouse muscle cell lines since GH is the principal regulator of IGF-I.
108 18772608 The MGF mRNA increased 1 h following GH stimulation whereas IGF-IEa mRNA, which encodes a systemic type of IGF-I, increased 4 h following GH stimulation.
109 18772608 Furthermore, GH induced the increase of MyoD as well as M-cadherin expression, the peak of which was parallel to that of MGF.
110 18772608 Mechano growth factor (MGF) is an alternatively spliced variant of insulin-like growth factor-I (IGF-I).
111 18772608 In the present study, we examined the effect of growth hormone (GH) on MGF expression in C2C12 mouse muscle cell lines since GH is the principal regulator of IGF-I.
112 18772608 The MGF mRNA increased 1 h following GH stimulation whereas IGF-IEa mRNA, which encodes a systemic type of IGF-I, increased 4 h following GH stimulation.
113 18772608 Furthermore, GH induced the increase of MyoD as well as M-cadherin expression, the peak of which was parallel to that of MGF.
114 18772608 Mechano growth factor (MGF) is an alternatively spliced variant of insulin-like growth factor-I (IGF-I).
115 18772608 In the present study, we examined the effect of growth hormone (GH) on MGF expression in C2C12 mouse muscle cell lines since GH is the principal regulator of IGF-I.
116 18772608 The MGF mRNA increased 1 h following GH stimulation whereas IGF-IEa mRNA, which encodes a systemic type of IGF-I, increased 4 h following GH stimulation.
117 18772608 Furthermore, GH induced the increase of MyoD as well as M-cadherin expression, the peak of which was parallel to that of MGF.
118 18772608 Mechano growth factor (MGF) is an alternatively spliced variant of insulin-like growth factor-I (IGF-I).
119 18772608 In the present study, we examined the effect of growth hormone (GH) on MGF expression in C2C12 mouse muscle cell lines since GH is the principal regulator of IGF-I.
120 18772608 The MGF mRNA increased 1 h following GH stimulation whereas IGF-IEa mRNA, which encodes a systemic type of IGF-I, increased 4 h following GH stimulation.
121 18772608 Furthermore, GH induced the increase of MyoD as well as M-cadherin expression, the peak of which was parallel to that of MGF.
122 19109928 Skp2 promotes adipocyte differentiation via a p27Kip1-independent mechanism in primary mouse embryonic fibroblasts.
123 19109928 Skp2, the substrate-binding subunit of an SCF ubiquitin ligase complex, is a key regulator of cell cycle progression that targets substrates for degradation by the 26S proteasome.
124 19109928 We have now shown that ablation of Skp2 in primary mouse embryonic fibroblasts (MEFs) results both in impairment of adipocyte differentiation and in the accumulation of the cyclin-dependent kinase inhibitor p27(Kip1), a principal target of the SCF(Skp2) complex.
125 19109928 Genetic ablation of p27(Kip1) in MEFs promoted both lipid accumulation and adipocyte-specific gene expression.
126 19109928 However, depletion of p27(Kip1) by adenovirus-mediated RNA interference failed to correct the impairment of adipocyte differentiation in Skp2(-/-) MEFs.
127 19109928 In contrast, troglitazone, a high-affinity ligand for peroxisome proliferator-activated receptor gamma (PPARgamma), largely restored lipid accumulation and PPARgamma gene expression in Skp2(-/-) MEFs.
128 19109928 Our data suggest that Skp2 plays an essential role in adipogenesis in MEFs in a manner that is at least in part independent of regulation of p27(Kip1) expression.
129 19109928 Skp2 promotes adipocyte differentiation via a p27Kip1-independent mechanism in primary mouse embryonic fibroblasts.
130 19109928 Skp2, the substrate-binding subunit of an SCF ubiquitin ligase complex, is a key regulator of cell cycle progression that targets substrates for degradation by the 26S proteasome.
131 19109928 We have now shown that ablation of Skp2 in primary mouse embryonic fibroblasts (MEFs) results both in impairment of adipocyte differentiation and in the accumulation of the cyclin-dependent kinase inhibitor p27(Kip1), a principal target of the SCF(Skp2) complex.
132 19109928 Genetic ablation of p27(Kip1) in MEFs promoted both lipid accumulation and adipocyte-specific gene expression.
133 19109928 However, depletion of p27(Kip1) by adenovirus-mediated RNA interference failed to correct the impairment of adipocyte differentiation in Skp2(-/-) MEFs.
134 19109928 In contrast, troglitazone, a high-affinity ligand for peroxisome proliferator-activated receptor gamma (PPARgamma), largely restored lipid accumulation and PPARgamma gene expression in Skp2(-/-) MEFs.
135 19109928 Our data suggest that Skp2 plays an essential role in adipogenesis in MEFs in a manner that is at least in part independent of regulation of p27(Kip1) expression.
136 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
137 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
138 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
139 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
140 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
141 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
142 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
143 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
144 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
145 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
146 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
147 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
148 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
149 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
150 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
151 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
152 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
153 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
154 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
155 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
156 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
157 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
158 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
159 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
160 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
161 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
162 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
163 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
164 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
165 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
166 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
167 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
168 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
169 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
170 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
171 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
172 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
173 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
174 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
175 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
176 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
177 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
178 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
179 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
180 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
181 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
182 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
183 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
184 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
185 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
186 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
187 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
188 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
189 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
190 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
191 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
192 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
193 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
194 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
195 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
196 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
197 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
198 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
199 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
200 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
201 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
202 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
203 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
204 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
205 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
206 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
207 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
208 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
209 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
210 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
211 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
212 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
213 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
214 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
215 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
216 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
217 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
218 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
219 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
220 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
221 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
222 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
223 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
224 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
225 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
226 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
227 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
228 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
229 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
230 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
231 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
232 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
233 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
234 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
235 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
236 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
237 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
238 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
239 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
240 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
241 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
242 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
243 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
244 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
245 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
246 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
247 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
248 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
249 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
250 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
251 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
252 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
253 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
254 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
255 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
256 19802467 Stem cell factor protects against neuronal apoptosis by activating AKT/ERK in diabetic mice.
257 19802467 Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF).
258 19802467 We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes.
259 19802467 The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL.
260 19802467 SCF, ERK1/2 and AKT were measured by Western blotting.
261 19802467 In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice.
262 19802467 The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 +/- 0.077%, 2.83 +/- 0.156%, 1.86 +/- 0.094%, and 1.78 +/- 0.095% (mean +/- SEM), respectively.
263 19802467 SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex.
264 19802467 Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex.
265 19802467 However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex.
266 19802467 The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways.
267 19802467 These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.
268 20476577 [The impact of diabetes mellitus and insulin interference on cortical SCF/KIT of mice].
269 20596251 Expression of stem cell factor/c-kit signaling pathway components in diabetic fibrovascular epiretinal membranes.
270 21233843 TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis.
271 21233843 Herein, we show that transforming growth factor β-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death.
272 21233843 Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade.
273 21233843 Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes.
274 21233843 Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis.
275 21233843 Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.
276 21233843 TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis.
277 21233843 Herein, we show that transforming growth factor β-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death.
278 21233843 Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade.
279 21233843 Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes.
280 21233843 Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis.
281 21233843 Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.
282 21233843 TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis.
283 21233843 Herein, we show that transforming growth factor β-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death.
284 21233843 Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade.
285 21233843 Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes.
286 21233843 Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis.
287 21233843 Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.
288 21233843 TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis.
289 21233843 Herein, we show that transforming growth factor β-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death.
290 21233843 Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade.
291 21233843 Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes.
292 21233843 Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis.
293 21233843 Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.
294 21233843 TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis.
295 21233843 Herein, we show that transforming growth factor β-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death.
296 21233843 Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade.
297 21233843 Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes.
298 21233843 Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis.
299 21233843 Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.
300 21233843 TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis.
301 21233843 Herein, we show that transforming growth factor β-activated kinase 1 (TAK1) directly regulates stem cell factor (SCF) expression, which activates the protein kinase B (PKB)α pro-survival pathway in a cell-autonomous manner to protect keratinocytes from ROS-mediated cell death.
302 21233843 Using organotypic skin co-culture and comparative growth factor array analysis, we revealed a cell-autonomous mechanism that involved the SCF/c-Kit/PKBα signaling cascade.
303 21233843 Ectopic expression of TAK1 or treatment with exogenous recombinant SCF restored the increased ROS production and apoptotic cell death in TAK1-deficient keratinocytes.
304 21233843 Conversely, normal keratinocytes treated with various inhibitors targeting the SCF/c-Kit/PKBα pathway exhibited increased ROS production and TNF-α- or anoikis-induced apoptosis.
305 21233843 Our study reveals a novel anti-apoptotic role for SCF in keratinocytes and identifies TAK1 as a novel player uniting inflammation and ROS regulation in skin redox biology.
306 21544097 Single 24 h transduction of human CD34+ cells with human immunodeficiency virus type 1 (HIV1)-based lentiviral vectors in media containing stem cell factor (SCF), FMS-like tyrosine kinase 3 (FLT3) ligand, thrombopoietin (each 100 ng ml⁻¹) and 10% fetal bovine serum was compared with various cytokine conditions during ex vivo culture and assayed using humanized xenograft mice for 6 months after transplantation.
307 21544097 In summary, 24 h prestimulation followed by single 24-h lentiviral transduction in serum-free media with SCF, FLT3 ligand and thrombopoietin yields high transduction efficiency to engrafting human CD34+ cells, and is applicable in human clinical gene therapy trials.
308 21544097 Single 24 h transduction of human CD34+ cells with human immunodeficiency virus type 1 (HIV1)-based lentiviral vectors in media containing stem cell factor (SCF), FMS-like tyrosine kinase 3 (FLT3) ligand, thrombopoietin (each 100 ng ml⁻¹) and 10% fetal bovine serum was compared with various cytokine conditions during ex vivo culture and assayed using humanized xenograft mice for 6 months after transplantation.
309 21544097 In summary, 24 h prestimulation followed by single 24-h lentiviral transduction in serum-free media with SCF, FLT3 ligand and thrombopoietin yields high transduction efficiency to engrafting human CD34+ cells, and is applicable in human clinical gene therapy trials.
310 22121118 To analyze these processes in greater detail, we have generated a Sox17-mCherry fusion (SCF) protein by gene targeting in ES cells.
311 22733969 These cells were generated from conventional NK cells by incubation with IL-18 and are characterized by the expression of the surface markers CD117 (also known as c-Kit, stem cell factor receptor) and programmed death (PD)-ligand 1.
312 22733969 In vitro analyses demonstrated a direct lysis activity of IL-18-stimulated NK cells against activated insulin-specific CD8(+) T cells in a PD-1/PD-ligand 1-dependent manner.
313 23269409 Downregulation of Fas activity rescues early onset of diabetes in c-Kit(Wv/+) mice.
314 23269409 c-Kit and its ligand stem cell factor (SCF) are important for β-cell survival and maturation; meanwhile, interactions between the Fas receptor (Fas) and Fas ligand are capable of triggering β-cell apoptosis.
315 23269409 Disruption of c-Kit signaling leads to severe loss of β-cell mass and function with upregulation of Fas expression in c-Kit(Wv/+) mouse islets, suggesting that there is a critical balance between c-Kit and Fas activation in β-cells.
316 23269409 In the present study, we investigated the interrelationship between c-Kit and Fas activation that mediates β-cell survival and function.
317 23269409 We generated double mutant, c-Kit(Wv/+);Fas(lpr/lpr) (Wv(-/-)), mice to study the physiological and functional role of Fas with respect to β-cell function in c-Kit(Wv/+) mice.
318 23269409 We observed that islets in c-Kit(Wv/+) mice showed a significant increase in β-cell apoptosis along with upregulated p53 and Fas expression.
319 23269409 These results were verified in vitro in INS-1 cells treated with SCF or c-Kit siRNA combined with a p53 inhibitor and Fas siRNA.
320 23269409 These findings demonstrate that a balance between the c-Kit and Fas signaling pathways is critical in the regulation of β-cell survival and function.
321 23269409 Downregulation of Fas activity rescues early onset of diabetes in c-Kit(Wv/+) mice.
322 23269409 c-Kit and its ligand stem cell factor (SCF) are important for β-cell survival and maturation; meanwhile, interactions between the Fas receptor (Fas) and Fas ligand are capable of triggering β-cell apoptosis.
323 23269409 Disruption of c-Kit signaling leads to severe loss of β-cell mass and function with upregulation of Fas expression in c-Kit(Wv/+) mouse islets, suggesting that there is a critical balance between c-Kit and Fas activation in β-cells.
324 23269409 In the present study, we investigated the interrelationship between c-Kit and Fas activation that mediates β-cell survival and function.
325 23269409 We generated double mutant, c-Kit(Wv/+);Fas(lpr/lpr) (Wv(-/-)), mice to study the physiological and functional role of Fas with respect to β-cell function in c-Kit(Wv/+) mice.
326 23269409 We observed that islets in c-Kit(Wv/+) mice showed a significant increase in β-cell apoptosis along with upregulated p53 and Fas expression.
327 23269409 These results were verified in vitro in INS-1 cells treated with SCF or c-Kit siRNA combined with a p53 inhibitor and Fas siRNA.
328 23269409 These findings demonstrate that a balance between the c-Kit and Fas signaling pathways is critical in the regulation of β-cell survival and function.
329 23352981 The results showed that the protein expression levels of c-Kit and membrane-bound stem cell factor (mSCF) in gastric smooth muscle layers were decreased in STZ-induced diabetic mice.
330 23352981 Pretreatment of the cultured gastric smooth muscle cells (GSMCs) with different concentration of CNP can significantly decrease the mSCF expression level. 8-Bromoguanosine-3',5'-cyclomo-nophosphate (8-Br-cGMP), a membrane permeable cGMP analog, mimicked the effect of CNP but not cANF (a specific NPR-C agonist).
331 23352981 These findings suggest that up-regulation of NPs/NPR-A, B/cGMP and NPs/NPR-C signaling pathways may be involved in diabetes-induced loss of gastric ICC.
332 23352981 The results showed that the protein expression levels of c-Kit and membrane-bound stem cell factor (mSCF) in gastric smooth muscle layers were decreased in STZ-induced diabetic mice.
333 23352981 Pretreatment of the cultured gastric smooth muscle cells (GSMCs) with different concentration of CNP can significantly decrease the mSCF expression level. 8-Bromoguanosine-3',5'-cyclomo-nophosphate (8-Br-cGMP), a membrane permeable cGMP analog, mimicked the effect of CNP but not cANF (a specific NPR-C agonist).
334 23352981 These findings suggest that up-regulation of NPs/NPR-A, B/cGMP and NPs/NPR-C signaling pathways may be involved in diabetes-induced loss of gastric ICC.
335 23448582 Curcumin improves expression of SCF/c-kit through attenuating oxidative stress and NF-κB activation in gastric tissues of diabetic gastroparesis rats.
336 23485602 Association of stem cell factor and high-sensitivity C reactive protein concentrations in crevicular fluid and serum in patients with chronic periodontitis with and without type 2 diabetes.
337 23485602 The aim of the present study was to clarify whether there is any correlation between the levels of high-sensitivity C reactive protein (hs-CRP) and stem cell factor (SCF) in serum and gingival crevicular fluid (GCF) of patients with chronic periodontitis (CP) with and without type 2 diabetes mellitus (DM).
338 23485602 Levels of hs-CRP and SCF in GCF and serum were quantified using different techniques.
339 23485602 Association of stem cell factor and high-sensitivity C reactive protein concentrations in crevicular fluid and serum in patients with chronic periodontitis with and without type 2 diabetes.
340 23485602 The aim of the present study was to clarify whether there is any correlation between the levels of high-sensitivity C reactive protein (hs-CRP) and stem cell factor (SCF) in serum and gingival crevicular fluid (GCF) of patients with chronic periodontitis (CP) with and without type 2 diabetes mellitus (DM).
341 23485602 Levels of hs-CRP and SCF in GCF and serum were quantified using different techniques.
342 23485602 Association of stem cell factor and high-sensitivity C reactive protein concentrations in crevicular fluid and serum in patients with chronic periodontitis with and without type 2 diabetes.
343 23485602 The aim of the present study was to clarify whether there is any correlation between the levels of high-sensitivity C reactive protein (hs-CRP) and stem cell factor (SCF) in serum and gingival crevicular fluid (GCF) of patients with chronic periodontitis (CP) with and without type 2 diabetes mellitus (DM).
344 23485602 Levels of hs-CRP and SCF in GCF and serum were quantified using different techniques.
345 23671874 Vascular endothelial growth factor (VEGF), soluble vascular endothelial growth factor receptor-2 (sVEGFR-2), stem cell factor (SCF), soluble c-kit (s-kit), endothelial nitric oxide synthase (eNOS), and prostaglandin E2 (PGE2) levels were measured by ELISA in vitreous samples from 34 PDR and 15 nondiabetic patients. eNOS was not detected.
346 23671874 VEGF, sVEGFR-2, SCF, and s-kit levels were significantly higher in PDR with active neovascularization compared with quiescent PDR and nondiabetic patients (P < 0.001; 0.007; 0.001; <0.001, resp.).
347 23671874 Our findings suggest that upregulation of VEGF, sVEGFR-2, SCF, and s-kit supports the contributions of angiogenesis and vasculogenesis in pathogenesis of PDR.
348 23671874 Vascular endothelial growth factor (VEGF), soluble vascular endothelial growth factor receptor-2 (sVEGFR-2), stem cell factor (SCF), soluble c-kit (s-kit), endothelial nitric oxide synthase (eNOS), and prostaglandin E2 (PGE2) levels were measured by ELISA in vitreous samples from 34 PDR and 15 nondiabetic patients. eNOS was not detected.
349 23671874 VEGF, sVEGFR-2, SCF, and s-kit levels were significantly higher in PDR with active neovascularization compared with quiescent PDR and nondiabetic patients (P < 0.001; 0.007; 0.001; <0.001, resp.).
350 23671874 Our findings suggest that upregulation of VEGF, sVEGFR-2, SCF, and s-kit supports the contributions of angiogenesis and vasculogenesis in pathogenesis of PDR.
351 23671874 Vascular endothelial growth factor (VEGF), soluble vascular endothelial growth factor receptor-2 (sVEGFR-2), stem cell factor (SCF), soluble c-kit (s-kit), endothelial nitric oxide synthase (eNOS), and prostaglandin E2 (PGE2) levels were measured by ELISA in vitreous samples from 34 PDR and 15 nondiabetic patients. eNOS was not detected.
352 23671874 VEGF, sVEGFR-2, SCF, and s-kit levels were significantly higher in PDR with active neovascularization compared with quiescent PDR and nondiabetic patients (P < 0.001; 0.007; 0.001; <0.001, resp.).
353 23671874 Our findings suggest that upregulation of VEGF, sVEGFR-2, SCF, and s-kit supports the contributions of angiogenesis and vasculogenesis in pathogenesis of PDR.
354 23745035 Effect of endogenous insulin-like growth factor and stem cell factor on diabetic colonic dysmotility.