anti gfp  (Cell Signaling Technology Inc)


Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc anti gfp
    Depletion <t>of</t> <t>Fign</t> improves functional recovery by increasing Tyr-tubulin. (A, B) Western blotting of tyrosinated (Tyr)-tubulin and Fign after spinal cord contusion (A) and sciatic nerve crush (B). α/β-Tubulin was used as a loading control. The data are shown as mean ± SE ( n = 3). The data were normalized by control (0 day) group. Linear regression showed expression trends of target proteins after injury. (C, D) Model of Fign depletion in spinal cord injury: <t>U6-GFP</t> Fign-AAV9 virus injected at T8, spinal cord contusion at T10 (C). Timeline of virus injection, spinal cord contusion, and Basso-Beattie-Bresnahan (BBB) evaluation (D). (E) BBB scores after spinal cord contusion (mean ± SE, n = 12 or 13). * P < 0.05; # P < 0.05, vs . Ctrl shRNA group (two-way analysis of variance followed by post hoc Bonferroni’s test). (F) Representative western blotting of Fign from injured spinal cord at 14 days after spinal cord contusion. α/β-Tubulin was used as a loading control (mean ± SE, n = 4). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. (G) Representative immunostaining results of NF200 (red, Cy3) in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Immunopositivity of NF200 was markedly increased in the injured area in the Fign shRNA group. Scale bars: 500 μm, 100 μm (enlarged images). (H) Statistical results of relative NF200 levels (mean ± SE, n = 8, Student’s t -test). (I) Representative immunostaining results of Tyr-tubulin and EB3 in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Microtubule plus-end binding protein EB3 significantly increased and mostly concentrated in Tyr-tubulin enriched areas in the Fign shRNA group. Tyr-tubulin is labeled in red (Alexa Fluor 647) and EB3 in yellow (Cy3) (changed to green by pseudo-color). Scale bars: 500 μm, 100 μm (enlarged images). (J) Statistical results of relative Tyr-tubulin and EB3 levels (mean ± SE, n = 5). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. AAV: Adeno-associated virus; BBB: Basso-Beattie-Bresnahan; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; NF200: neurofilament-200; SCI: spinal cord injury; shRNA: short hairpin RNA; Tyr-tubulin: tyrosinated-tubulin.
    Anti Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/anti gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    anti gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "Fidgetin interacting with microtubule end binding protein EB3 affects axonal regrowth in spinal cord injury"

    Article Title: Fidgetin interacting with microtubule end binding protein EB3 affects axonal regrowth in spinal cord injury

    Journal: Neural Regeneration Research

    doi: 10.4103/1673-5374.373716

    Depletion of Fign improves functional recovery by increasing Tyr-tubulin. (A, B) Western blotting of tyrosinated (Tyr)-tubulin and Fign after spinal cord contusion (A) and sciatic nerve crush (B). α/β-Tubulin was used as a loading control. The data are shown as mean ± SE ( n = 3). The data were normalized by control (0 day) group. Linear regression showed expression trends of target proteins after injury. (C, D) Model of Fign depletion in spinal cord injury: U6-GFP Fign-AAV9 virus injected at T8, spinal cord contusion at T10 (C). Timeline of virus injection, spinal cord contusion, and Basso-Beattie-Bresnahan (BBB) evaluation (D). (E) BBB scores after spinal cord contusion (mean ± SE, n = 12 or 13). * P < 0.05; # P < 0.05, vs . Ctrl shRNA group (two-way analysis of variance followed by post hoc Bonferroni’s test). (F) Representative western blotting of Fign from injured spinal cord at 14 days after spinal cord contusion. α/β-Tubulin was used as a loading control (mean ± SE, n = 4). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. (G) Representative immunostaining results of NF200 (red, Cy3) in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Immunopositivity of NF200 was markedly increased in the injured area in the Fign shRNA group. Scale bars: 500 μm, 100 μm (enlarged images). (H) Statistical results of relative NF200 levels (mean ± SE, n = 8, Student’s t -test). (I) Representative immunostaining results of Tyr-tubulin and EB3 in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Microtubule plus-end binding protein EB3 significantly increased and mostly concentrated in Tyr-tubulin enriched areas in the Fign shRNA group. Tyr-tubulin is labeled in red (Alexa Fluor 647) and EB3 in yellow (Cy3) (changed to green by pseudo-color). Scale bars: 500 μm, 100 μm (enlarged images). (J) Statistical results of relative Tyr-tubulin and EB3 levels (mean ± SE, n = 5). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. AAV: Adeno-associated virus; BBB: Basso-Beattie-Bresnahan; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; NF200: neurofilament-200; SCI: spinal cord injury; shRNA: short hairpin RNA; Tyr-tubulin: tyrosinated-tubulin.
    Figure Legend Snippet: Depletion of Fign improves functional recovery by increasing Tyr-tubulin. (A, B) Western blotting of tyrosinated (Tyr)-tubulin and Fign after spinal cord contusion (A) and sciatic nerve crush (B). α/β-Tubulin was used as a loading control. The data are shown as mean ± SE ( n = 3). The data were normalized by control (0 day) group. Linear regression showed expression trends of target proteins after injury. (C, D) Model of Fign depletion in spinal cord injury: U6-GFP Fign-AAV9 virus injected at T8, spinal cord contusion at T10 (C). Timeline of virus injection, spinal cord contusion, and Basso-Beattie-Bresnahan (BBB) evaluation (D). (E) BBB scores after spinal cord contusion (mean ± SE, n = 12 or 13). * P < 0.05; # P < 0.05, vs . Ctrl shRNA group (two-way analysis of variance followed by post hoc Bonferroni’s test). (F) Representative western blotting of Fign from injured spinal cord at 14 days after spinal cord contusion. α/β-Tubulin was used as a loading control (mean ± SE, n = 4). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. (G) Representative immunostaining results of NF200 (red, Cy3) in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Immunopositivity of NF200 was markedly increased in the injured area in the Fign shRNA group. Scale bars: 500 μm, 100 μm (enlarged images). (H) Statistical results of relative NF200 levels (mean ± SE, n = 8, Student’s t -test). (I) Representative immunostaining results of Tyr-tubulin and EB3 in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Microtubule plus-end binding protein EB3 significantly increased and mostly concentrated in Tyr-tubulin enriched areas in the Fign shRNA group. Tyr-tubulin is labeled in red (Alexa Fluor 647) and EB3 in yellow (Cy3) (changed to green by pseudo-color). Scale bars: 500 μm, 100 μm (enlarged images). (J) Statistical results of relative Tyr-tubulin and EB3 levels (mean ± SE, n = 5). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. AAV: Adeno-associated virus; BBB: Basso-Beattie-Bresnahan; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; NF200: neurofilament-200; SCI: spinal cord injury; shRNA: short hairpin RNA; Tyr-tubulin: tyrosinated-tubulin.

    Techniques Used: Functional Assay, Western Blot, Expressing, Injection, shRNA, Immunostaining, Binding Assay, Labeling

    Fign regulates Tyr-tubulin by binding to EB3 and parking at microtubules plus-ends. (A) Representative inverted immunostaining of Fign (green, Alexa Fluor 488), tyrosinated (Tyr)-tubulin (red, Cy3), and actin (violet, Alexa Fluor 647). Scale bars: 20 μm, 5 μm (enlarged images). (B) Representative western blotting with GFP antibody after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. (C) Representative western blotting of tubulin proteins after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. The data (normalized by control GFP plasmid transfection) are shown as mean ± SE ( n = 3), and were analyzed by Student’s t -test. (D) Representative inverted immunostaining of Tyr-tubulin (red, Cy3) and acetylated-tubulin (green, Alexa Fluor 488) after E18 cortical neurons were transfected with Ctrl or EB3 siRNA and lentivirus (LV)5-NC or LV5-Fign. Ctrl siRNA combined with LV5-Fign decreased Tyr-tubulin and axonal length. EB3 siRNA combined with LV5-Fign rescued this phenomenon. Scale bar: 20 μm. (E) Statistical results of relative Tyr-tubulin levels (left panel, n = 15, normalized by Ctrl siRNA + LV5-NC group) and axonal length (right panel, n = 35) of neurons transfected with Ctrl or EB3 siRNA, together with LV5-NC or LV5-Fign. The data are shown as mean ± SE and were analyzed using one-way analysis of variance and Dunnett’s test. (F) Representative inverted immunostaining of Fign in E18 cortical neurons transfected with LV5-NC or LV5-Fign. Scale bar: 20 μm. Red dotted box shows Fign in the axon. Red arrow shows the axonal terminal. Ace-tubulin: Acetylated-tubulin; E18: rat embryo at 18 days; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; LV: lentivirus; P: pellet, tubulins in polymerized microtubules; S: supernatant, soluble tubulins; siRNA: small interfering RNA; T: total, total tubulins; Tyr-tubulin: tyrosinated-tubulin.
    Figure Legend Snippet: Fign regulates Tyr-tubulin by binding to EB3 and parking at microtubules plus-ends. (A) Representative inverted immunostaining of Fign (green, Alexa Fluor 488), tyrosinated (Tyr)-tubulin (red, Cy3), and actin (violet, Alexa Fluor 647). Scale bars: 20 μm, 5 μm (enlarged images). (B) Representative western blotting with GFP antibody after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. (C) Representative western blotting of tubulin proteins after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. The data (normalized by control GFP plasmid transfection) are shown as mean ± SE ( n = 3), and were analyzed by Student’s t -test. (D) Representative inverted immunostaining of Tyr-tubulin (red, Cy3) and acetylated-tubulin (green, Alexa Fluor 488) after E18 cortical neurons were transfected with Ctrl or EB3 siRNA and lentivirus (LV)5-NC or LV5-Fign. Ctrl siRNA combined with LV5-Fign decreased Tyr-tubulin and axonal length. EB3 siRNA combined with LV5-Fign rescued this phenomenon. Scale bar: 20 μm. (E) Statistical results of relative Tyr-tubulin levels (left panel, n = 15, normalized by Ctrl siRNA + LV5-NC group) and axonal length (right panel, n = 35) of neurons transfected with Ctrl or EB3 siRNA, together with LV5-NC or LV5-Fign. The data are shown as mean ± SE and were analyzed using one-way analysis of variance and Dunnett’s test. (F) Representative inverted immunostaining of Fign in E18 cortical neurons transfected with LV5-NC or LV5-Fign. Scale bar: 20 μm. Red dotted box shows Fign in the axon. Red arrow shows the axonal terminal. Ace-tubulin: Acetylated-tubulin; E18: rat embryo at 18 days; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; LV: lentivirus; P: pellet, tubulins in polymerized microtubules; S: supernatant, soluble tubulins; siRNA: small interfering RNA; T: total, total tubulins; Tyr-tubulin: tyrosinated-tubulin.

    Techniques Used: Binding Assay, Immunostaining, Western Blot, Transfection, Plasmid Preparation, Small Interfering RNA

    gfp  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc gfp
    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing <t>luciferase-GFP</t> were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.
    Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance"

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    Journal: Nature cancer

    doi: 10.1038/s43018-023-00553-8

    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.
    Figure Legend Snippet: a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.

    Techniques Used: Expressing, Luciferase, Injection, Flow Cytometry, Two Tailed Test

    a, Representative Haematoxylin and eosin (H&E) (top) and Masson’s trichrome (bottom) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors from 2-3 independent experiments. Scale bars, 100μm. b, Immunohistochemical (IHC) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage of SA-β-gal+ area and the number of Ki67+ and pRb+ cells per field are shown inset (n=2-4 per group). Scale bar, 50μm. c, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors grown in different organs and treated as in (b). Quantification of the percentage of GFP+ (green) tumor cells expressing p21 (cyan) is shown inset (n=2-4 per group). Scale bar, 50μm. d, GFP+ tumor cells were FACS sorted from indicated tumors and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). Gene Set Enrichment Analysis (GSEA) of RNA-seq data using an established senescence gene set is shown. NES, normalized enrichment score. P values in d were calculated using two-sided, Kolmogorov-Smirnov test. Error bars, mean ± SEM.
    Figure Legend Snippet: a, Representative Haematoxylin and eosin (H&E) (top) and Masson’s trichrome (bottom) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors from 2-3 independent experiments. Scale bars, 100μm. b, Immunohistochemical (IHC) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage of SA-β-gal+ area and the number of Ki67+ and pRb+ cells per field are shown inset (n=2-4 per group). Scale bar, 50μm. c, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors grown in different organs and treated as in (b). Quantification of the percentage of GFP+ (green) tumor cells expressing p21 (cyan) is shown inset (n=2-4 per group). Scale bar, 50μm. d, GFP+ tumor cells were FACS sorted from indicated tumors and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). Gene Set Enrichment Analysis (GSEA) of RNA-seq data using an established senescence gene set is shown. NES, normalized enrichment score. P values in d were calculated using two-sided, Kolmogorov-Smirnov test. Error bars, mean ± SEM.

    Techniques Used: In Vivo, Staining, Derivative Assay, Immunohistochemistry, Immunofluorescence, Expressing, RNA Sequencing Assay

    a, KPC1 PDAC or KP1 LUAD tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. GFP+ tumor cells were FACS sorted and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). b, KEGG pathway analysis of pathways enriched in tumors in the lungs (LIL, PIL) compared to tumors in the pancreas (PIP, LIP) following T/P treatment. c, Heatmap showing fold change in SASP gene expression following T/P treatment in indicated tumor settings. d, Fold change in expression of select SASP chemokines following T/P treatment in indicated tumor settings (n=2-4 per group). Error bars, mean ± SEM. e, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumors in the pancreas (PIP, LIP) following T/P treatment. f, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. g, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL) and KP1 LUAD (LIL, LIP) derived-tumors grown in different organs and treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (PIP V; PIP TP; PIL TP; LIL TP and LIP TP, n=3, PIL V; LIL V and LIP V, n=2 independent tumors). Scale bar, 50μm. P values in b were calculated using two-sided, hypergeometric test and those in f using two-sided, Kolmogorov-Smirnov test.
    Figure Legend Snippet: a, KPC1 PDAC or KP1 LUAD tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. GFP+ tumor cells were FACS sorted and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). b, KEGG pathway analysis of pathways enriched in tumors in the lungs (LIL, PIL) compared to tumors in the pancreas (PIP, LIP) following T/P treatment. c, Heatmap showing fold change in SASP gene expression following T/P treatment in indicated tumor settings. d, Fold change in expression of select SASP chemokines following T/P treatment in indicated tumor settings (n=2-4 per group). Error bars, mean ± SEM. e, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumors in the pancreas (PIP, LIP) following T/P treatment. f, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. g, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL) and KP1 LUAD (LIL, LIP) derived-tumors grown in different organs and treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (PIP V; PIP TP; PIL TP; LIL TP and LIP TP, n=3, PIL V; LIL V and LIP V, n=2 independent tumors). Scale bar, 50μm. P values in b were calculated using two-sided, hypergeometric test and those in f using two-sided, Kolmogorov-Smirnov test.

    Techniques Used: Expressing, Luciferase, Injection, RNA Sequencing Assay, Immunofluorescence, Staining, Derivative Assay

    a, Schematic of KPC PDAC syngeneic orthotopic transplantation into 8-16 week old male and female SMA-TK mice and treatment regimens. b, Immunohistochemical (IHC) staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated with vehicle, trametinib (1 mg/kg) and palbociclib (100 mg/kg), and/or ganciclovir (GCV) (50 mg/kg) for 2 weeks. Quantification of number of SMA+ cells per field is shown inset (b, V, n=5; TP, n=4; GCV, n=4; and TP/GCV, n=5 independent tumors). Scale bar, 50μm. c, Waterfall plot of the response of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice to treatment as in (b) (c, V, n=6; TP, n=10; GCV, n=8, and TP/GCV, n=15 independent mice). Data represents pool of 2 independent experiments. d-e, Flow cytometry analysis of NK (d) and T cell (e) numbers and activation markers in KPC1 orthotopic PDAC tumors propagated in SMA-TK mice following treatment as in (b) (n=8-16 per group). (d-e, V, n=8; TP, n=9; GCV, n=8, and TP/GCV, n=16 independent mice). Data represents pool of 2 independent experiments. Error bars, mean ± SEM. f, KEGG (left) and REACTOME (right) pathway analysis of RNA-seq data generated from FACS sorted GFP+ tumor cells from SMA-TK mice harboring KPC1 orthotopic PDAC tumors and treated as in (b) (n=4-5 per group). g, Heatmap of RNA-seq analysis of SASP gene expression in PDAC cells from KPC1 orthotopic PDAC tumors propagated in SMA-TK mice and treated as in (b) (n=4-5 per group). h, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumor cells from KPC1 orthotopic PDAC propagated in SMA-TK mice treated with T/P alone compared with combined T/P/GCV treatment. i, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. j, Immunofluorescence staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated as in (b). Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (n=2 per group). Scale bar, 50μm. P values in c-e were calculated using two-tailed, unpaired Student’s t-test, f using two-sided, hypergeometric test, and i using two-sided, Kolmogorov-Smirnov test.
    Figure Legend Snippet: a, Schematic of KPC PDAC syngeneic orthotopic transplantation into 8-16 week old male and female SMA-TK mice and treatment regimens. b, Immunohistochemical (IHC) staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated with vehicle, trametinib (1 mg/kg) and palbociclib (100 mg/kg), and/or ganciclovir (GCV) (50 mg/kg) for 2 weeks. Quantification of number of SMA+ cells per field is shown inset (b, V, n=5; TP, n=4; GCV, n=4; and TP/GCV, n=5 independent tumors). Scale bar, 50μm. c, Waterfall plot of the response of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice to treatment as in (b) (c, V, n=6; TP, n=10; GCV, n=8, and TP/GCV, n=15 independent mice). Data represents pool of 2 independent experiments. d-e, Flow cytometry analysis of NK (d) and T cell (e) numbers and activation markers in KPC1 orthotopic PDAC tumors propagated in SMA-TK mice following treatment as in (b) (n=8-16 per group). (d-e, V, n=8; TP, n=9; GCV, n=8, and TP/GCV, n=16 independent mice). Data represents pool of 2 independent experiments. Error bars, mean ± SEM. f, KEGG (left) and REACTOME (right) pathway analysis of RNA-seq data generated from FACS sorted GFP+ tumor cells from SMA-TK mice harboring KPC1 orthotopic PDAC tumors and treated as in (b) (n=4-5 per group). g, Heatmap of RNA-seq analysis of SASP gene expression in PDAC cells from KPC1 orthotopic PDAC tumors propagated in SMA-TK mice and treated as in (b) (n=4-5 per group). h, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumor cells from KPC1 orthotopic PDAC propagated in SMA-TK mice treated with T/P alone compared with combined T/P/GCV treatment. i, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. j, Immunofluorescence staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated as in (b). Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (n=2 per group). Scale bar, 50μm. P values in c-e were calculated using two-tailed, unpaired Student’s t-test, f using two-sided, hypergeometric test, and i using two-sided, Kolmogorov-Smirnov test.

    Techniques Used: Transplantation Assay, Immunohistochemistry, Flow Cytometry, Activation Assay, RNA Sequencing Assay, Generated, Expressing, Immunofluorescence, Staining, Two Tailed Test

    a-b, KPC2 PDAC or KP2 LUAD tumor cells expressing GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Flow cytometry analysis of NK cell numbers and degranulation in PDAC (PIP, PIL) (a) and LUAD-derived tumors (LIL, LIP) (b) grown in different organs are shown (a-b, PIP V, n=3; PIP TP, n=4; PIL V and PIL TP, n=8 independent mice). c, Flow cytometry analysis of NK cell numbers and degranulation in spleens of mice with KPC1-derived PIP tumors treated as in (a) (n=5 independent mice per group). d, Kaplan-Meier survival curve of mice with KPC2-derived PIP tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) (d, V, n=5; TP; TP+αNK1.1 and TP+αCD8, n=8). e, IVIS images showing luciferase signaling in KPC1-derived PIL tumors following treatment as in (a). Right, quantification of total luminescence in the thoracic region (e, V, n=5; TP and TP+αNK1.1, n=8 independent mice). f, Waterfall plot of the response of KPC1-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 depleting antibody (PK136; 250 μg) (f, V and TP, n=5, TP+αNK1.1, n=6 independent mice). g, Waterfall plot of the response of KPC2-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) depleting antibody (g, V, n=5; TP, n=7; TP+αNK1.1 and TP+αCD8 ,n=8 independent mice). h, Kaplan-Meier survival curve of mice with KPC1-derived PIL tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against CD8 (2.43; 200 μg) or CD4 (GK1.5; 200 μg) (h, V, n=5; TP, TP+αNK1.1 and TP+αCD8, n=7 independent mice). i, Flow cytometry analysis of CD4+ and CD8+ T cell numbers and degranulation in KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD-derived tumors (LIL, LIP, LILiver) grown in different organs and treated as in (a) (i, PIP V, PIP TP, PIL TP, n=10; PIL V, n=9; LIL V, n=3; LIL TP, n=5; LIP V, n=13; LIP TP, n=15; PILiver V, n=9; PILiver TP, n=10; LILiver V, n=8; LILiver TP, n=10 independent mice). Data represents pool of 3 independent experiments. P values in a-c, e-g, and i were calculated using two-tailed, unpaired Student’s t-test, and those in d and h were calculated using log-rank test. Error bars, mean ± SEM.
    Figure Legend Snippet: a-b, KPC2 PDAC or KP2 LUAD tumor cells expressing GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Flow cytometry analysis of NK cell numbers and degranulation in PDAC (PIP, PIL) (a) and LUAD-derived tumors (LIL, LIP) (b) grown in different organs are shown (a-b, PIP V, n=3; PIP TP, n=4; PIL V and PIL TP, n=8 independent mice). c, Flow cytometry analysis of NK cell numbers and degranulation in spleens of mice with KPC1-derived PIP tumors treated as in (a) (n=5 independent mice per group). d, Kaplan-Meier survival curve of mice with KPC2-derived PIP tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) (d, V, n=5; TP; TP+αNK1.1 and TP+αCD8, n=8). e, IVIS images showing luciferase signaling in KPC1-derived PIL tumors following treatment as in (a). Right, quantification of total luminescence in the thoracic region (e, V, n=5; TP and TP+αNK1.1, n=8 independent mice). f, Waterfall plot of the response of KPC1-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 depleting antibody (PK136; 250 μg) (f, V and TP, n=5, TP+αNK1.1, n=6 independent mice). g, Waterfall plot of the response of KPC2-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) depleting antibody (g, V, n=5; TP, n=7; TP+αNK1.1 and TP+αCD8 ,n=8 independent mice). h, Kaplan-Meier survival curve of mice with KPC1-derived PIL tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against CD8 (2.43; 200 μg) or CD4 (GK1.5; 200 μg) (h, V, n=5; TP, TP+αNK1.1 and TP+αCD8, n=7 independent mice). i, Flow cytometry analysis of CD4+ and CD8+ T cell numbers and degranulation in KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD-derived tumors (LIL, LIP, LILiver) grown in different organs and treated as in (a) (i, PIP V, PIP TP, PIL TP, n=10; PIL V, n=9; LIL V, n=3; LIL TP, n=5; LIP V, n=13; LIP TP, n=15; PILiver V, n=9; PILiver TP, n=10; LILiver V, n=8; LILiver TP, n=10 independent mice). Data represents pool of 3 independent experiments. P values in a-c, e-g, and i were calculated using two-tailed, unpaired Student’s t-test, and those in d and h were calculated using log-rank test. Error bars, mean ± SEM.

    Techniques Used: Expressing, Injection, Flow Cytometry, Derivative Assay, Luciferase, Two Tailed Test

    anti gfp antibody  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc anti gfp antibody
    Anti Gfp Antibody, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/anti gfp antibody/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    anti gfp antibody - by Bioz Stars, 2023-09
    86/100 stars

    Images

    gfp  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc gfp
    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing <t>luciferase-GFP</t> were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.
    Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance"

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    Journal: Nature cancer

    doi: 10.1038/s43018-023-00553-8

    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.
    Figure Legend Snippet: a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.

    Techniques Used: Expressing, Luciferase, Injection, Flow Cytometry, Two Tailed Test

    a, Representative Haematoxylin and eosin (H&E) (top) and Masson’s trichrome (bottom) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors from 2-3 independent experiments. Scale bars, 100μm. b, Immunohistochemical (IHC) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage of SA-β-gal+ area and the number of Ki67+ and pRb+ cells per field are shown inset (n=2-4 per group). Scale bar, 50μm. c, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors grown in different organs and treated as in (b). Quantification of the percentage of GFP+ (green) tumor cells expressing p21 (cyan) is shown inset (n=2-4 per group). Scale bar, 50μm. d, GFP+ tumor cells were FACS sorted from indicated tumors and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). Gene Set Enrichment Analysis (GSEA) of RNA-seq data using an established senescence gene set is shown. NES, normalized enrichment score. P values in d were calculated using two-sided, Kolmogorov-Smirnov test. Error bars, mean ± SEM.
    Figure Legend Snippet: a, Representative Haematoxylin and eosin (H&E) (top) and Masson’s trichrome (bottom) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors from 2-3 independent experiments. Scale bars, 100μm. b, Immunohistochemical (IHC) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage of SA-β-gal+ area and the number of Ki67+ and pRb+ cells per field are shown inset (n=2-4 per group). Scale bar, 50μm. c, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors grown in different organs and treated as in (b). Quantification of the percentage of GFP+ (green) tumor cells expressing p21 (cyan) is shown inset (n=2-4 per group). Scale bar, 50μm. d, GFP+ tumor cells were FACS sorted from indicated tumors and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). Gene Set Enrichment Analysis (GSEA) of RNA-seq data using an established senescence gene set is shown. NES, normalized enrichment score. P values in d were calculated using two-sided, Kolmogorov-Smirnov test. Error bars, mean ± SEM.

    Techniques Used: In Vivo, Staining, Derivative Assay, Immunohistochemistry, Immunofluorescence, Expressing, RNA Sequencing Assay

    a, KPC1 PDAC or KP1 LUAD tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. GFP+ tumor cells were FACS sorted and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). b, KEGG pathway analysis of pathways enriched in tumors in the lungs (LIL, PIL) compared to tumors in the pancreas (PIP, LIP) following T/P treatment. c, Heatmap showing fold change in SASP gene expression following T/P treatment in indicated tumor settings. d, Fold change in expression of select SASP chemokines following T/P treatment in indicated tumor settings (n=2-4 per group). Error bars, mean ± SEM. e, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumors in the pancreas (PIP, LIP) following T/P treatment. f, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. g, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL) and KP1 LUAD (LIL, LIP) derived-tumors grown in different organs and treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (PIP V; PIP TP; PIL TP; LIL TP and LIP TP, n=3, PIL V; LIL V and LIP V, n=2 independent tumors). Scale bar, 50μm. P values in b were calculated using two-sided, hypergeometric test and those in f using two-sided, Kolmogorov-Smirnov test.
    Figure Legend Snippet: a, KPC1 PDAC or KP1 LUAD tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. GFP+ tumor cells were FACS sorted and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). b, KEGG pathway analysis of pathways enriched in tumors in the lungs (LIL, PIL) compared to tumors in the pancreas (PIP, LIP) following T/P treatment. c, Heatmap showing fold change in SASP gene expression following T/P treatment in indicated tumor settings. d, Fold change in expression of select SASP chemokines following T/P treatment in indicated tumor settings (n=2-4 per group). Error bars, mean ± SEM. e, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumors in the pancreas (PIP, LIP) following T/P treatment. f, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. g, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL) and KP1 LUAD (LIL, LIP) derived-tumors grown in different organs and treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (PIP V; PIP TP; PIL TP; LIL TP and LIP TP, n=3, PIL V; LIL V and LIP V, n=2 independent tumors). Scale bar, 50μm. P values in b were calculated using two-sided, hypergeometric test and those in f using two-sided, Kolmogorov-Smirnov test.

    Techniques Used: Expressing, Luciferase, Injection, RNA Sequencing Assay, Immunofluorescence, Staining, Derivative Assay

    a, Schematic of KPC PDAC syngeneic orthotopic transplantation into 8-16 week old male and female SMA-TK mice and treatment regimens. b, Immunohistochemical (IHC) staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated with vehicle, trametinib (1 mg/kg) and palbociclib (100 mg/kg), and/or ganciclovir (GCV) (50 mg/kg) for 2 weeks. Quantification of number of SMA+ cells per field is shown inset (b, V, n=5; TP, n=4; GCV, n=4; and TP/GCV, n=5 independent tumors). Scale bar, 50μm. c, Waterfall plot of the response of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice to treatment as in (b) (c, V, n=6; TP, n=10; GCV, n=8, and TP/GCV, n=15 independent mice). Data represents pool of 2 independent experiments. d-e, Flow cytometry analysis of NK (d) and T cell (e) numbers and activation markers in KPC1 orthotopic PDAC tumors propagated in SMA-TK mice following treatment as in (b) (n=8-16 per group). (d-e, V, n=8; TP, n=9; GCV, n=8, and TP/GCV, n=16 independent mice). Data represents pool of 2 independent experiments. Error bars, mean ± SEM. f, KEGG (left) and REACTOME (right) pathway analysis of RNA-seq data generated from FACS sorted GFP+ tumor cells from SMA-TK mice harboring KPC1 orthotopic PDAC tumors and treated as in (b) (n=4-5 per group). g, Heatmap of RNA-seq analysis of SASP gene expression in PDAC cells from KPC1 orthotopic PDAC tumors propagated in SMA-TK mice and treated as in (b) (n=4-5 per group). h, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumor cells from KPC1 orthotopic PDAC propagated in SMA-TK mice treated with T/P alone compared with combined T/P/GCV treatment. i, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. j, Immunofluorescence staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated as in (b). Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (n=2 per group). Scale bar, 50μm. P values in c-e were calculated using two-tailed, unpaired Student’s t-test, f using two-sided, hypergeometric test, and i using two-sided, Kolmogorov-Smirnov test.
    Figure Legend Snippet: a, Schematic of KPC PDAC syngeneic orthotopic transplantation into 8-16 week old male and female SMA-TK mice and treatment regimens. b, Immunohistochemical (IHC) staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated with vehicle, trametinib (1 mg/kg) and palbociclib (100 mg/kg), and/or ganciclovir (GCV) (50 mg/kg) for 2 weeks. Quantification of number of SMA+ cells per field is shown inset (b, V, n=5; TP, n=4; GCV, n=4; and TP/GCV, n=5 independent tumors). Scale bar, 50μm. c, Waterfall plot of the response of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice to treatment as in (b) (c, V, n=6; TP, n=10; GCV, n=8, and TP/GCV, n=15 independent mice). Data represents pool of 2 independent experiments. d-e, Flow cytometry analysis of NK (d) and T cell (e) numbers and activation markers in KPC1 orthotopic PDAC tumors propagated in SMA-TK mice following treatment as in (b) (n=8-16 per group). (d-e, V, n=8; TP, n=9; GCV, n=8, and TP/GCV, n=16 independent mice). Data represents pool of 2 independent experiments. Error bars, mean ± SEM. f, KEGG (left) and REACTOME (right) pathway analysis of RNA-seq data generated from FACS sorted GFP+ tumor cells from SMA-TK mice harboring KPC1 orthotopic PDAC tumors and treated as in (b) (n=4-5 per group). g, Heatmap of RNA-seq analysis of SASP gene expression in PDAC cells from KPC1 orthotopic PDAC tumors propagated in SMA-TK mice and treated as in (b) (n=4-5 per group). h, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumor cells from KPC1 orthotopic PDAC propagated in SMA-TK mice treated with T/P alone compared with combined T/P/GCV treatment. i, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. j, Immunofluorescence staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated as in (b). Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (n=2 per group). Scale bar, 50μm. P values in c-e were calculated using two-tailed, unpaired Student’s t-test, f using two-sided, hypergeometric test, and i using two-sided, Kolmogorov-Smirnov test.

    Techniques Used: Transplantation Assay, Immunohistochemistry, Flow Cytometry, Activation Assay, RNA Sequencing Assay, Generated, Expressing, Immunofluorescence, Staining, Two Tailed Test

    a-b, KPC2 PDAC or KP2 LUAD tumor cells expressing GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Flow cytometry analysis of NK cell numbers and degranulation in PDAC (PIP, PIL) (a) and LUAD-derived tumors (LIL, LIP) (b) grown in different organs are shown (a-b, PIP V, n=3; PIP TP, n=4; PIL V and PIL TP, n=8 independent mice). c, Flow cytometry analysis of NK cell numbers and degranulation in spleens of mice with KPC1-derived PIP tumors treated as in (a) (n=5 independent mice per group). d, Kaplan-Meier survival curve of mice with KPC2-derived PIP tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) (d, V, n=5; TP; TP+αNK1.1 and TP+αCD8, n=8). e, IVIS images showing luciferase signaling in KPC1-derived PIL tumors following treatment as in (a). Right, quantification of total luminescence in the thoracic region (e, V, n=5; TP and TP+αNK1.1, n=8 independent mice). f, Waterfall plot of the response of KPC1-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 depleting antibody (PK136; 250 μg) (f, V and TP, n=5, TP+αNK1.1, n=6 independent mice). g, Waterfall plot of the response of KPC2-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) depleting antibody (g, V, n=5; TP, n=7; TP+αNK1.1 and TP+αCD8 ,n=8 independent mice). h, Kaplan-Meier survival curve of mice with KPC1-derived PIL tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against CD8 (2.43; 200 μg) or CD4 (GK1.5; 200 μg) (h, V, n=5; TP, TP+αNK1.1 and TP+αCD8, n=7 independent mice). i, Flow cytometry analysis of CD4+ and CD8+ T cell numbers and degranulation in KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD-derived tumors (LIL, LIP, LILiver) grown in different organs and treated as in (a) (i, PIP V, PIP TP, PIL TP, n=10; PIL V, n=9; LIL V, n=3; LIL TP, n=5; LIP V, n=13; LIP TP, n=15; PILiver V, n=9; PILiver TP, n=10; LILiver V, n=8; LILiver TP, n=10 independent mice). Data represents pool of 3 independent experiments. P values in a-c, e-g, and i were calculated using two-tailed, unpaired Student’s t-test, and those in d and h were calculated using log-rank test. Error bars, mean ± SEM.
    Figure Legend Snippet: a-b, KPC2 PDAC or KP2 LUAD tumor cells expressing GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Flow cytometry analysis of NK cell numbers and degranulation in PDAC (PIP, PIL) (a) and LUAD-derived tumors (LIL, LIP) (b) grown in different organs are shown (a-b, PIP V, n=3; PIP TP, n=4; PIL V and PIL TP, n=8 independent mice). c, Flow cytometry analysis of NK cell numbers and degranulation in spleens of mice with KPC1-derived PIP tumors treated as in (a) (n=5 independent mice per group). d, Kaplan-Meier survival curve of mice with KPC2-derived PIP tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) (d, V, n=5; TP; TP+αNK1.1 and TP+αCD8, n=8). e, IVIS images showing luciferase signaling in KPC1-derived PIL tumors following treatment as in (a). Right, quantification of total luminescence in the thoracic region (e, V, n=5; TP and TP+αNK1.1, n=8 independent mice). f, Waterfall plot of the response of KPC1-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 depleting antibody (PK136; 250 μg) (f, V and TP, n=5, TP+αNK1.1, n=6 independent mice). g, Waterfall plot of the response of KPC2-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) depleting antibody (g, V, n=5; TP, n=7; TP+αNK1.1 and TP+αCD8 ,n=8 independent mice). h, Kaplan-Meier survival curve of mice with KPC1-derived PIL tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against CD8 (2.43; 200 μg) or CD4 (GK1.5; 200 μg) (h, V, n=5; TP, TP+αNK1.1 and TP+αCD8, n=7 independent mice). i, Flow cytometry analysis of CD4+ and CD8+ T cell numbers and degranulation in KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD-derived tumors (LIL, LIP, LILiver) grown in different organs and treated as in (a) (i, PIP V, PIP TP, PIL TP, n=10; PIL V, n=9; LIL V, n=3; LIL TP, n=5; LIP V, n=13; LIP TP, n=15; PILiver V, n=9; PILiver TP, n=10; LILiver V, n=8; LILiver TP, n=10 independent mice). Data represents pool of 3 independent experiments. P values in a-c, e-g, and i were calculated using two-tailed, unpaired Student’s t-test, and those in d and h were calculated using log-rank test. Error bars, mean ± SEM.

    Techniques Used: Expressing, Injection, Flow Cytometry, Derivative Assay, Luciferase, Two Tailed Test

    chicken anti gfp  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc chicken anti gfp
    (A) Accessibility profiles of astrocyte-specific enhancers in the human whole-body accessibility atlas . Single-cell profiles were grouped within each tissue into pseudo-bulk aggregates, then normalized according to the signal (reads in peaks) within the dataset. Accessibility profiles are likely to predict enhancer activities within each tissue. Focusing on liver, some astrocyte-specific enhancers are predicted to have high expression, and some are predicted to have very little or no expression. In contrast, accessibility atlases do not predict expression of GFAP promoter across tissues. (B) Whole livers from mice injected intravenously with eHGT_381h- and eHGT_390m-enhancer-AAV vectors, stained <t>with</t> <t>anti-GFP</t> antibody. eHGT_381h has high liver accessibility, is predicted to have high liver expression, and shows many strong SYFP2-expressing hepatocytes throughout the liver as predicted. In contrast, eHGT_390m has very little liver accessibility and so is predicted to have little liver expression, and in fact shows few positive SYFP2-expressing hepatocytes as predicted. (C) Agreement between liver expression predictions and liver expression measurements across several astrocyte-specific enhancer-AAV vectors. eHGT_371m, 371h, 381h, and 386m all show many SYFP2-expressing hepatocytes as predicted. eHGT_390m, 390h, 375m, and 387m show few weak SYFP2-expressing hepatocytes as predicted. GFAP promoter shows many expressing hepatocytes, which was not predictable from the accessibility atlases. eHGT_380h shows many SYFP2-expressing astrocytes, in contrast to the epigenetic prediction. Liver images in B and C represent one to two mice analyzed for each vector. (D-E) Testing fidelity of enhancer-AAV expression across disease states. We used a Dravet syndrome model Scn1a R613X/+ mouse to induce epilepsy-associated hippocampal gliosis, injected enhancer-AAVs prior to the critical period, and analyzed tissue for expression patterns after the critical period (D). We assessed hippocampal gliosis with anti-GFAP antibody and enhancer-AAV expression <t>with</t> <t>anti-GFP</t> antibody (E). eHGT_390m maintained specific expression and similar levels in hippocampal astrocytes regardless of epileptic gliosis. In contrast, GFAP promoter expression strongly increased in gliotic astrocytes, and also was observed in dentate gyrus granule cells. Red dashed rectangles indicate the position of the expanded zoomed view, and the curved arrows indicate a rotated view. Abbreviations: ML molecular layer, GCL granule cell layer, PoL polymorphic layer.
    Chicken Anti Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/chicken anti gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    chicken anti gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "Enhancer-AAVs allow genetic access to oligodendrocytes and diverse populations of astrocytes across species"

    Article Title: Enhancer-AAVs allow genetic access to oligodendrocytes and diverse populations of astrocytes across species

    Journal: bioRxiv

    doi: 10.1101/2023.09.20.558718

    (A) Accessibility profiles of astrocyte-specific enhancers in the human whole-body accessibility atlas . Single-cell profiles were grouped within each tissue into pseudo-bulk aggregates, then normalized according to the signal (reads in peaks) within the dataset. Accessibility profiles are likely to predict enhancer activities within each tissue. Focusing on liver, some astrocyte-specific enhancers are predicted to have high expression, and some are predicted to have very little or no expression. In contrast, accessibility atlases do not predict expression of GFAP promoter across tissues. (B) Whole livers from mice injected intravenously with eHGT_381h- and eHGT_390m-enhancer-AAV vectors, stained with anti-GFP antibody. eHGT_381h has high liver accessibility, is predicted to have high liver expression, and shows many strong SYFP2-expressing hepatocytes throughout the liver as predicted. In contrast, eHGT_390m has very little liver accessibility and so is predicted to have little liver expression, and in fact shows few positive SYFP2-expressing hepatocytes as predicted. (C) Agreement between liver expression predictions and liver expression measurements across several astrocyte-specific enhancer-AAV vectors. eHGT_371m, 371h, 381h, and 386m all show many SYFP2-expressing hepatocytes as predicted. eHGT_390m, 390h, 375m, and 387m show few weak SYFP2-expressing hepatocytes as predicted. GFAP promoter shows many expressing hepatocytes, which was not predictable from the accessibility atlases. eHGT_380h shows many SYFP2-expressing astrocytes, in contrast to the epigenetic prediction. Liver images in B and C represent one to two mice analyzed for each vector. (D-E) Testing fidelity of enhancer-AAV expression across disease states. We used a Dravet syndrome model Scn1a R613X/+ mouse to induce epilepsy-associated hippocampal gliosis, injected enhancer-AAVs prior to the critical period, and analyzed tissue for expression patterns after the critical period (D). We assessed hippocampal gliosis with anti-GFAP antibody and enhancer-AAV expression with anti-GFP antibody (E). eHGT_390m maintained specific expression and similar levels in hippocampal astrocytes regardless of epileptic gliosis. In contrast, GFAP promoter expression strongly increased in gliotic astrocytes, and also was observed in dentate gyrus granule cells. Red dashed rectangles indicate the position of the expanded zoomed view, and the curved arrows indicate a rotated view. Abbreviations: ML molecular layer, GCL granule cell layer, PoL polymorphic layer.
    Figure Legend Snippet: (A) Accessibility profiles of astrocyte-specific enhancers in the human whole-body accessibility atlas . Single-cell profiles were grouped within each tissue into pseudo-bulk aggregates, then normalized according to the signal (reads in peaks) within the dataset. Accessibility profiles are likely to predict enhancer activities within each tissue. Focusing on liver, some astrocyte-specific enhancers are predicted to have high expression, and some are predicted to have very little or no expression. In contrast, accessibility atlases do not predict expression of GFAP promoter across tissues. (B) Whole livers from mice injected intravenously with eHGT_381h- and eHGT_390m-enhancer-AAV vectors, stained with anti-GFP antibody. eHGT_381h has high liver accessibility, is predicted to have high liver expression, and shows many strong SYFP2-expressing hepatocytes throughout the liver as predicted. In contrast, eHGT_390m has very little liver accessibility and so is predicted to have little liver expression, and in fact shows few positive SYFP2-expressing hepatocytes as predicted. (C) Agreement between liver expression predictions and liver expression measurements across several astrocyte-specific enhancer-AAV vectors. eHGT_371m, 371h, 381h, and 386m all show many SYFP2-expressing hepatocytes as predicted. eHGT_390m, 390h, 375m, and 387m show few weak SYFP2-expressing hepatocytes as predicted. GFAP promoter shows many expressing hepatocytes, which was not predictable from the accessibility atlases. eHGT_380h shows many SYFP2-expressing astrocytes, in contrast to the epigenetic prediction. Liver images in B and C represent one to two mice analyzed for each vector. (D-E) Testing fidelity of enhancer-AAV expression across disease states. We used a Dravet syndrome model Scn1a R613X/+ mouse to induce epilepsy-associated hippocampal gliosis, injected enhancer-AAVs prior to the critical period, and analyzed tissue for expression patterns after the critical period (D). We assessed hippocampal gliosis with anti-GFAP antibody and enhancer-AAV expression with anti-GFP antibody (E). eHGT_390m maintained specific expression and similar levels in hippocampal astrocytes regardless of epileptic gliosis. In contrast, GFAP promoter expression strongly increased in gliotic astrocytes, and also was observed in dentate gyrus granule cells. Red dashed rectangles indicate the position of the expanded zoomed view, and the curved arrows indicate a rotated view. Abbreviations: ML molecular layer, GCL granule cell layer, PoL polymorphic layer.

    Techniques Used: Expressing, Injection, Staining, Plasmid Preparation

    gfp  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc gfp
    Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    rabbit anti gfp antibody  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc rabbit anti gfp antibody
    A. Left: Consensus motif of KEAP1 KELCH as established with the HD2 library and the aligned SQSTM1 peptide with the P348L mutation site indicated in blue. Right: Displacement curves from KEAP1 KELCH domain with wild-type/P348L SQSTM1 334-349 peptide. Measurements were in at least technical triplets. B. <t>GFP</t> trap of YFP or GFPKEAP1 and probing for co-immunoprecipitation of wild-type/P348L Myc-SQSTM1 in HeLa cells (n=3). C-D . Network of KEAP1 ( C ) or KPNA4 ( D ) with the prey proteins from the domain-mutation pairs. Interactions and corresponding mutations disrupted by the mutation are indicated in blue, enhanced in red and neutral effects in grey. A green dot indicates previously reported interacting prey proteins and a straight line if the domain’s binding motif is found in the binding peptide. The circle size encodes the confidence level of the domain-mutation pairs. For KPNA4 the network has been reduced to include the reported interactors and the domain-mutation pairs with an associated p-value ≤ 0.001. E. Left: Consensus motif of KPNA4 ARM as established with the HD2 library and the aligned CDC45 and ABRAXAS1 peptides with the mutation sites indicated in blue. Right: Displacement curves from KPNA4 ARM domain with wild-type/R157C CDC45 152-166 and wild-type/R361Q ABRAXAS1 349-364 peptides. Measurements were in technical triplets. F-G. Representative images of the localisation of wild-type and mutant EGFP-tagged (R361Q) ABRAXAS1 and (R157C) CDC45 in relation to the nuclear Hoechst staining (n=3). H. GFP trap of EGFP (n=3), wildtype EGFP-CDC45 (n=3) or mutant R157C EGFP-CDC45 (n=2) and probing for co-immunoprecipitation <t>of</t> <t>T7-KPNA7</t> in HEK293T cells.
    Rabbit Anti Gfp Antibody, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/rabbit anti gfp antibody/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    rabbit anti gfp antibody - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "Proteome-scale characterisation of protein motif interactome rewiring by disease mutations"

    Article Title: Proteome-scale characterisation of protein motif interactome rewiring by disease mutations

    Journal: bioRxiv

    doi: 10.1101/2023.09.18.558189

    A. Left: Consensus motif of KEAP1 KELCH as established with the HD2 library and the aligned SQSTM1 peptide with the P348L mutation site indicated in blue. Right: Displacement curves from KEAP1 KELCH domain with wild-type/P348L SQSTM1 334-349 peptide. Measurements were in at least technical triplets. B. GFP trap of YFP or GFPKEAP1 and probing for co-immunoprecipitation of wild-type/P348L Myc-SQSTM1 in HeLa cells (n=3). C-D . Network of KEAP1 ( C ) or KPNA4 ( D ) with the prey proteins from the domain-mutation pairs. Interactions and corresponding mutations disrupted by the mutation are indicated in blue, enhanced in red and neutral effects in grey. A green dot indicates previously reported interacting prey proteins and a straight line if the domain’s binding motif is found in the binding peptide. The circle size encodes the confidence level of the domain-mutation pairs. For KPNA4 the network has been reduced to include the reported interactors and the domain-mutation pairs with an associated p-value ≤ 0.001. E. Left: Consensus motif of KPNA4 ARM as established with the HD2 library and the aligned CDC45 and ABRAXAS1 peptides with the mutation sites indicated in blue. Right: Displacement curves from KPNA4 ARM domain with wild-type/R157C CDC45 152-166 and wild-type/R361Q ABRAXAS1 349-364 peptides. Measurements were in technical triplets. F-G. Representative images of the localisation of wild-type and mutant EGFP-tagged (R361Q) ABRAXAS1 and (R157C) CDC45 in relation to the nuclear Hoechst staining (n=3). H. GFP trap of EGFP (n=3), wildtype EGFP-CDC45 (n=3) or mutant R157C EGFP-CDC45 (n=2) and probing for co-immunoprecipitation of T7-KPNA7 in HEK293T cells.
    Figure Legend Snippet: A. Left: Consensus motif of KEAP1 KELCH as established with the HD2 library and the aligned SQSTM1 peptide with the P348L mutation site indicated in blue. Right: Displacement curves from KEAP1 KELCH domain with wild-type/P348L SQSTM1 334-349 peptide. Measurements were in at least technical triplets. B. GFP trap of YFP or GFPKEAP1 and probing for co-immunoprecipitation of wild-type/P348L Myc-SQSTM1 in HeLa cells (n=3). C-D . Network of KEAP1 ( C ) or KPNA4 ( D ) with the prey proteins from the domain-mutation pairs. Interactions and corresponding mutations disrupted by the mutation are indicated in blue, enhanced in red and neutral effects in grey. A green dot indicates previously reported interacting prey proteins and a straight line if the domain’s binding motif is found in the binding peptide. The circle size encodes the confidence level of the domain-mutation pairs. For KPNA4 the network has been reduced to include the reported interactors and the domain-mutation pairs with an associated p-value ≤ 0.001. E. Left: Consensus motif of KPNA4 ARM as established with the HD2 library and the aligned CDC45 and ABRAXAS1 peptides with the mutation sites indicated in blue. Right: Displacement curves from KPNA4 ARM domain with wild-type/R157C CDC45 152-166 and wild-type/R361Q ABRAXAS1 349-364 peptides. Measurements were in technical triplets. F-G. Representative images of the localisation of wild-type and mutant EGFP-tagged (R361Q) ABRAXAS1 and (R157C) CDC45 in relation to the nuclear Hoechst staining (n=3). H. GFP trap of EGFP (n=3), wildtype EGFP-CDC45 (n=3) or mutant R157C EGFP-CDC45 (n=2) and probing for co-immunoprecipitation of T7-KPNA7 in HEK293T cells.

    Techniques Used: Mutagenesis, Immunoprecipitation, Binding Assay, Staining

    gfp  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc gfp
    (A) Scheme for expression of Cp OGA WT or Cp OGA DM in various Drosophila brain structures using different Gal4 drivers. (B) Immunostaining of adult Drosophila brains. Brains were stained with anti-O-GlcNAc antibody RL2 (red) to assess O-GlcNAcylation level, <t>and</t> <t>anti-GFP</t> (green) antibody to validate tissue-specific expression of Cp OGA. Nuclei were stained with DAPI (blue). Scale bar: 100 μm. (C) Quantification of fluorescent intensity of O-GlcNAc staining in Cp OGA WT or Cp OGA DM expressed brains. (D) Immunostaining of adult Drosophila brains. Outlined areas indicate the cell bodies of kenyon cells in mushroom body. Scale bar: 100 μm. (E) Quantification of relative fluorescent intensity of O-GlcNAc staining in Cp OGA WT or Cp OGA DM expressed brain structures. (F) A compilation of performance index in learning test of the indicated flies expressing either Cp OGA WT or Cp OGA DM . (G) A compilation of learing performance index of flies expressing Cp OGA WT or Cp OGA DM only in the mushroom body at adult stage. Each datapoint represents an independent experiment with approximately 200 flies. p values were determined by unpaired t -test, and the stars indicate significant differences (*** p < 0.001, ** p < 0.01 and ns, not significant, p ≥ 0.05). Error bars represent SD. —source data 1. Excel spreadsheet containing source data used to generate .
    Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "Olfactory learning in Drosophila requires O-GlcNAcylation of mushroom body ribosomal subunits"

    Article Title: Olfactory learning in Drosophila requires O-GlcNAcylation of mushroom body ribosomal subunits

    Journal: bioRxiv

    doi: 10.1101/2023.09.14.557796

    (A) Scheme for expression of Cp OGA WT or Cp OGA DM in various Drosophila brain structures using different Gal4 drivers. (B) Immunostaining of adult Drosophila brains. Brains were stained with anti-O-GlcNAc antibody RL2 (red) to assess O-GlcNAcylation level, and anti-GFP (green) antibody to validate tissue-specific expression of Cp OGA. Nuclei were stained with DAPI (blue). Scale bar: 100 μm. (C) Quantification of fluorescent intensity of O-GlcNAc staining in Cp OGA WT or Cp OGA DM expressed brains. (D) Immunostaining of adult Drosophila brains. Outlined areas indicate the cell bodies of kenyon cells in mushroom body. Scale bar: 100 μm. (E) Quantification of relative fluorescent intensity of O-GlcNAc staining in Cp OGA WT or Cp OGA DM expressed brain structures. (F) A compilation of performance index in learning test of the indicated flies expressing either Cp OGA WT or Cp OGA DM . (G) A compilation of learing performance index of flies expressing Cp OGA WT or Cp OGA DM only in the mushroom body at adult stage. Each datapoint represents an independent experiment with approximately 200 flies. p values were determined by unpaired t -test, and the stars indicate significant differences (*** p < 0.001, ** p < 0.01 and ns, not significant, p ≥ 0.05). Error bars represent SD. —source data 1. Excel spreadsheet containing source data used to generate .
    Figure Legend Snippet: (A) Scheme for expression of Cp OGA WT or Cp OGA DM in various Drosophila brain structures using different Gal4 drivers. (B) Immunostaining of adult Drosophila brains. Brains were stained with anti-O-GlcNAc antibody RL2 (red) to assess O-GlcNAcylation level, and anti-GFP (green) antibody to validate tissue-specific expression of Cp OGA. Nuclei were stained with DAPI (blue). Scale bar: 100 μm. (C) Quantification of fluorescent intensity of O-GlcNAc staining in Cp OGA WT or Cp OGA DM expressed brains. (D) Immunostaining of adult Drosophila brains. Outlined areas indicate the cell bodies of kenyon cells in mushroom body. Scale bar: 100 μm. (E) Quantification of relative fluorescent intensity of O-GlcNAc staining in Cp OGA WT or Cp OGA DM expressed brain structures. (F) A compilation of performance index in learning test of the indicated flies expressing either Cp OGA WT or Cp OGA DM . (G) A compilation of learing performance index of flies expressing Cp OGA WT or Cp OGA DM only in the mushroom body at adult stage. Each datapoint represents an independent experiment with approximately 200 flies. p values were determined by unpaired t -test, and the stars indicate significant differences (*** p < 0.001, ** p < 0.01 and ns, not significant, p ≥ 0.05). Error bars represent SD. —source data 1. Excel spreadsheet containing source data used to generate .

    Techniques Used: Expressing, Immunostaining, Staining

    (A) Heatmaps showing the mRNA levels (upper) and the normalized O-GlcNAc levels (lower) of the identified ribosomal candidates in different brain regions. (B) Immunoprecipitation of ribosomes using FLAG-tagged RpL13A. The expression of RpL13A-FLAG was validated by immunoblotting with anti-FLAG antibody. Ribosomal proteins were visualized using silver staining, and O-GlcNAcylation of ribosomes was analyzed by immunoblotting with anti-O-GlcNAc antibody RL2. (C) A compilation of performance index of the indicated flies in the learning test. Learning defect of flies expressing Cp OGA WT was corrected by selective expression of dMyc in mushroom body. Each datapoint represents an independent experiment with approximately 200 flies. (D) Ex vivo measurement of protein synthesis in mushroom body using the OPP assay. Brains from the indicated flies were stained with anti-GFP (green) antibody to validate Cp OGA expression, and OPP (grey) to quantify protein synthesis. Nuclei were visualized with DAPI (blue). Outlined areas indicate the cell bodies of kenyon cells of mushroom body. Scale bar: 100 μm. (E) Quantification of relative OPP fluorescent intensity in mushroom body regions. p values were determined by unpaired t -test, the stars indicate significant differences (*** p < 0.001, ** p < 0.01, * p < 0.05). Error bars represent SD. —source data 1. Raw data of all western blots from . —source data 2. Complete and uncropped membranes of all western blots from . —source data 3. Excel spreadsheet containing source data used to generate .
    Figure Legend Snippet: (A) Heatmaps showing the mRNA levels (upper) and the normalized O-GlcNAc levels (lower) of the identified ribosomal candidates in different brain regions. (B) Immunoprecipitation of ribosomes using FLAG-tagged RpL13A. The expression of RpL13A-FLAG was validated by immunoblotting with anti-FLAG antibody. Ribosomal proteins were visualized using silver staining, and O-GlcNAcylation of ribosomes was analyzed by immunoblotting with anti-O-GlcNAc antibody RL2. (C) A compilation of performance index of the indicated flies in the learning test. Learning defect of flies expressing Cp OGA WT was corrected by selective expression of dMyc in mushroom body. Each datapoint represents an independent experiment with approximately 200 flies. (D) Ex vivo measurement of protein synthesis in mushroom body using the OPP assay. Brains from the indicated flies were stained with anti-GFP (green) antibody to validate Cp OGA expression, and OPP (grey) to quantify protein synthesis. Nuclei were visualized with DAPI (blue). Outlined areas indicate the cell bodies of kenyon cells of mushroom body. Scale bar: 100 μm. (E) Quantification of relative OPP fluorescent intensity in mushroom body regions. p values were determined by unpaired t -test, the stars indicate significant differences (*** p < 0.001, ** p < 0.01, * p < 0.05). Error bars represent SD. —source data 1. Raw data of all western blots from . —source data 2. Complete and uncropped membranes of all western blots from . —source data 3. Excel spreadsheet containing source data used to generate .

    Techniques Used: Immunoprecipitation, Expressing, Western Blot, Silver Staining, Ex Vivo, Staining

    anti gfp  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc anti gfp
    a Knockdown of Sirt1 activates NF-κB and inhibits autophagy in endothelial cells. Immunoblotting and quantification of p-p65/p65, p62, and LC3 II/LC3 I in HUVECs transfected with control siRNA ( n = 5 independent experiments, gray bar) vs. Sirt1 siRNA ( n = 5 independent experiments, blue bar); representative immunoblots are shown . b Overexpression of p65 upregulates endothelial Tβ4 mRNA. Immunoblotting of Flag-tagged p65 and qPCR of Tβ4 mRNA in HUVECs (scramble control, n = 5 independent experiments, gray bar) vs. Flag-p65 ( n = 3 independent experiments, yellow bar). P = 0.008; representative immunoblots are shown . c–f Inhibition of NF-κB negates Tβ4 upregulation in Sirt1-deficient endothelial cells. c Immunoblotting of Sirt1, p65, and p-p65; representative immunoblots are shown, and ( d ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 7 independent experiments, gray bar), Sirt1 siRNA ( n = 7 independent experiments, blue bar), treated with Bay 11-7082 only ( n = 7 independent experiments, yellow bar), and Sirt1 siRNA + Bay 11-7082 ( n = 5 independent experiments, light blue bar); ( e ) Immunoblotting of Sirt1, <t>IκBα-GFP,</t> p65, and p-p65, and GAPDH; representative immunoblots are shown, and ( f ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 6 independent experiments, gray bar), Sirt1 siRNA ( n = 6 independent experiments, blue bar), IκBα-GFP only ( n = 6 independent experiments, yellow bar), and Sirt1-si+ IκBα-GFP ( n = 8 independent experiments, light blue bar). g–k Inhibition of autophagy upregulates endothelial Tβ4, and restoration of autophagy negates Tβ4 upregulation in Sirt1-deficient endothelial cells. g Immunoblotting and quantification of Atg7, Sirt1, p62, LC3, p65, and p-p65; representative immunoblots are shown. Gray bar is control siRNA ( n = 6 independent experiments), and blue bar is Atg7-si ( n = 6 independent experiments); and ( h ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 9 independent experiments, gray bar) or Atg7 siRNA ( n = 9 independent experiments, blue bar). i Immunoblotting of Sirt1, Atg3, LC3; and j qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 8 independent experiments, gray bar) or Sirt1 siRNA ( n = 8 independent experiments, blue bar), mCherry-Atg3 only ( n = 8 independent experiments, light blue bar), and Sirt1-si+mCherry-Atg3 ( n = 10 independent experiments, brown bar); representative immunoblots are shown . k Overexpression of p62 upregulates Tβ4. Immunoblotting of HA and p62, and qPCR of Tβ4 mRNA in HUVECs transfected with HA-tagged p62 ( n = 6 independent experiments, yellow bar) and scramble control ( n = 4 independent experiments, gray bar); representative immunoblots are shown. Data are shown as mean ± SEM. Two-tailed unpaired Student’s t test was used. Each dot in the bar figures represents one sample. C-si scramble siRNA, Sirt1-si Sirt1 siRNA, Atg7-si Atg7 siRNA, p-p65 phosphorylation p65, Sirt1 fl/fl Sirt1 flox/flox mice, KO E-Sirt1-KO mice. Source data are provided as a Source Data file. l Summary of the mechanisms underlying enhanced insulin sensitivity in mice deficient of endothelial Sirt1. Deficiency of endothelial Sirt1 impairs autophagy and stimulates NF-κB signaling to boost Tβ4 expression and secretion that <t>enhances</t> <t>ILK-mediated</t> Akt activity in skeletal muscle cells.
    Anti Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/anti gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    anti gfp - by Bioz Stars, 2023-09
    86/100 stars

    Images

    1) Product Images from "Deficiency of endothelial sirtuin1 in mice stimulates skeletal muscle insulin sensitivity by modifying the secretome"

    Article Title: Deficiency of endothelial sirtuin1 in mice stimulates skeletal muscle insulin sensitivity by modifying the secretome

    Journal: Nature Communications

    doi: 10.1038/s41467-023-41351-1

    a Knockdown of Sirt1 activates NF-κB and inhibits autophagy in endothelial cells. Immunoblotting and quantification of p-p65/p65, p62, and LC3 II/LC3 I in HUVECs transfected with control siRNA ( n = 5 independent experiments, gray bar) vs. Sirt1 siRNA ( n = 5 independent experiments, blue bar); representative immunoblots are shown . b Overexpression of p65 upregulates endothelial Tβ4 mRNA. Immunoblotting of Flag-tagged p65 and qPCR of Tβ4 mRNA in HUVECs (scramble control, n = 5 independent experiments, gray bar) vs. Flag-p65 ( n = 3 independent experiments, yellow bar). P = 0.008; representative immunoblots are shown . c–f Inhibition of NF-κB negates Tβ4 upregulation in Sirt1-deficient endothelial cells. c Immunoblotting of Sirt1, p65, and p-p65; representative immunoblots are shown, and ( d ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 7 independent experiments, gray bar), Sirt1 siRNA ( n = 7 independent experiments, blue bar), treated with Bay 11-7082 only ( n = 7 independent experiments, yellow bar), and Sirt1 siRNA + Bay 11-7082 ( n = 5 independent experiments, light blue bar); ( e ) Immunoblotting of Sirt1, IκBα-GFP, p65, and p-p65, and GAPDH; representative immunoblots are shown, and ( f ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 6 independent experiments, gray bar), Sirt1 siRNA ( n = 6 independent experiments, blue bar), IκBα-GFP only ( n = 6 independent experiments, yellow bar), and Sirt1-si+ IκBα-GFP ( n = 8 independent experiments, light blue bar). g–k Inhibition of autophagy upregulates endothelial Tβ4, and restoration of autophagy negates Tβ4 upregulation in Sirt1-deficient endothelial cells. g Immunoblotting and quantification of Atg7, Sirt1, p62, LC3, p65, and p-p65; representative immunoblots are shown. Gray bar is control siRNA ( n = 6 independent experiments), and blue bar is Atg7-si ( n = 6 independent experiments); and ( h ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 9 independent experiments, gray bar) or Atg7 siRNA ( n = 9 independent experiments, blue bar). i Immunoblotting of Sirt1, Atg3, LC3; and j qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 8 independent experiments, gray bar) or Sirt1 siRNA ( n = 8 independent experiments, blue bar), mCherry-Atg3 only ( n = 8 independent experiments, light blue bar), and Sirt1-si+mCherry-Atg3 ( n = 10 independent experiments, brown bar); representative immunoblots are shown . k Overexpression of p62 upregulates Tβ4. Immunoblotting of HA and p62, and qPCR of Tβ4 mRNA in HUVECs transfected with HA-tagged p62 ( n = 6 independent experiments, yellow bar) and scramble control ( n = 4 independent experiments, gray bar); representative immunoblots are shown. Data are shown as mean ± SEM. Two-tailed unpaired Student’s t test was used. Each dot in the bar figures represents one sample. C-si scramble siRNA, Sirt1-si Sirt1 siRNA, Atg7-si Atg7 siRNA, p-p65 phosphorylation p65, Sirt1 fl/fl Sirt1 flox/flox mice, KO E-Sirt1-KO mice. Source data are provided as a Source Data file. l Summary of the mechanisms underlying enhanced insulin sensitivity in mice deficient of endothelial Sirt1. Deficiency of endothelial Sirt1 impairs autophagy and stimulates NF-κB signaling to boost Tβ4 expression and secretion that enhances ILK-mediated Akt activity in skeletal muscle cells.
    Figure Legend Snippet: a Knockdown of Sirt1 activates NF-κB and inhibits autophagy in endothelial cells. Immunoblotting and quantification of p-p65/p65, p62, and LC3 II/LC3 I in HUVECs transfected with control siRNA ( n = 5 independent experiments, gray bar) vs. Sirt1 siRNA ( n = 5 independent experiments, blue bar); representative immunoblots are shown . b Overexpression of p65 upregulates endothelial Tβ4 mRNA. Immunoblotting of Flag-tagged p65 and qPCR of Tβ4 mRNA in HUVECs (scramble control, n = 5 independent experiments, gray bar) vs. Flag-p65 ( n = 3 independent experiments, yellow bar). P = 0.008; representative immunoblots are shown . c–f Inhibition of NF-κB negates Tβ4 upregulation in Sirt1-deficient endothelial cells. c Immunoblotting of Sirt1, p65, and p-p65; representative immunoblots are shown, and ( d ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 7 independent experiments, gray bar), Sirt1 siRNA ( n = 7 independent experiments, blue bar), treated with Bay 11-7082 only ( n = 7 independent experiments, yellow bar), and Sirt1 siRNA + Bay 11-7082 ( n = 5 independent experiments, light blue bar); ( e ) Immunoblotting of Sirt1, IκBα-GFP, p65, and p-p65, and GAPDH; representative immunoblots are shown, and ( f ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 6 independent experiments, gray bar), Sirt1 siRNA ( n = 6 independent experiments, blue bar), IκBα-GFP only ( n = 6 independent experiments, yellow bar), and Sirt1-si+ IκBα-GFP ( n = 8 independent experiments, light blue bar). g–k Inhibition of autophagy upregulates endothelial Tβ4, and restoration of autophagy negates Tβ4 upregulation in Sirt1-deficient endothelial cells. g Immunoblotting and quantification of Atg7, Sirt1, p62, LC3, p65, and p-p65; representative immunoblots are shown. Gray bar is control siRNA ( n = 6 independent experiments), and blue bar is Atg7-si ( n = 6 independent experiments); and ( h ) qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 9 independent experiments, gray bar) or Atg7 siRNA ( n = 9 independent experiments, blue bar). i Immunoblotting of Sirt1, Atg3, LC3; and j qPCR of Tβ4 mRNA in HUVECs transfected with control siRNA ( n = 8 independent experiments, gray bar) or Sirt1 siRNA ( n = 8 independent experiments, blue bar), mCherry-Atg3 only ( n = 8 independent experiments, light blue bar), and Sirt1-si+mCherry-Atg3 ( n = 10 independent experiments, brown bar); representative immunoblots are shown . k Overexpression of p62 upregulates Tβ4. Immunoblotting of HA and p62, and qPCR of Tβ4 mRNA in HUVECs transfected with HA-tagged p62 ( n = 6 independent experiments, yellow bar) and scramble control ( n = 4 independent experiments, gray bar); representative immunoblots are shown. Data are shown as mean ± SEM. Two-tailed unpaired Student’s t test was used. Each dot in the bar figures represents one sample. C-si scramble siRNA, Sirt1-si Sirt1 siRNA, Atg7-si Atg7 siRNA, p-p65 phosphorylation p65, Sirt1 fl/fl Sirt1 flox/flox mice, KO E-Sirt1-KO mice. Source data are provided as a Source Data file. l Summary of the mechanisms underlying enhanced insulin sensitivity in mice deficient of endothelial Sirt1. Deficiency of endothelial Sirt1 impairs autophagy and stimulates NF-κB signaling to boost Tβ4 expression and secretion that enhances ILK-mediated Akt activity in skeletal muscle cells.

    Techniques Used: Western Blot, Transfection, Over Expression, Inhibition, Two Tailed Test, Expressing, Activity Assay

    anti gfp antibody  (Cell Signaling Technology Inc)


    Bioz Verified Symbol Cell Signaling Technology Inc is a verified supplier
    Bioz Manufacturer Symbol Cell Signaling Technology Inc manufactures this product  
  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86

    Structured Review

    Cell Signaling Technology Inc anti gfp antibody
    Anti Gfp Antibody, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/anti gfp antibody/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    anti gfp antibody - by Bioz Stars, 2023-09
    86/100 stars

    Images

    Similar Products

  • Logo
  • About
  • News
  • Press Release
  • Team
  • Advisors
  • Partners
  • Contact
  • Bioz Stars
  • Bioz vStars
  • 86
    Cell Signaling Technology Inc anti gfp
    Depletion <t>of</t> <t>Fign</t> improves functional recovery by increasing Tyr-tubulin. (A, B) Western blotting of tyrosinated (Tyr)-tubulin and Fign after spinal cord contusion (A) and sciatic nerve crush (B). α/β-Tubulin was used as a loading control. The data are shown as mean ± SE ( n = 3). The data were normalized by control (0 day) group. Linear regression showed expression trends of target proteins after injury. (C, D) Model of Fign depletion in spinal cord injury: <t>U6-GFP</t> Fign-AAV9 virus injected at T8, spinal cord contusion at T10 (C). Timeline of virus injection, spinal cord contusion, and Basso-Beattie-Bresnahan (BBB) evaluation (D). (E) BBB scores after spinal cord contusion (mean ± SE, n = 12 or 13). * P < 0.05; # P < 0.05, vs . Ctrl shRNA group (two-way analysis of variance followed by post hoc Bonferroni’s test). (F) Representative western blotting of Fign from injured spinal cord at 14 days after spinal cord contusion. α/β-Tubulin was used as a loading control (mean ± SE, n = 4). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. (G) Representative immunostaining results of NF200 (red, Cy3) in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Immunopositivity of NF200 was markedly increased in the injured area in the Fign shRNA group. Scale bars: 500 μm, 100 μm (enlarged images). (H) Statistical results of relative NF200 levels (mean ± SE, n = 8, Student’s t -test). (I) Representative immunostaining results of Tyr-tubulin and EB3 in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Microtubule plus-end binding protein EB3 significantly increased and mostly concentrated in Tyr-tubulin enriched areas in the Fign shRNA group. Tyr-tubulin is labeled in red (Alexa Fluor 647) and EB3 in yellow (Cy3) (changed to green by pseudo-color). Scale bars: 500 μm, 100 μm (enlarged images). (J) Statistical results of relative Tyr-tubulin and EB3 levels (mean ± SE, n = 5). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. AAV: Adeno-associated virus; BBB: Basso-Beattie-Bresnahan; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; NF200: neurofilament-200; SCI: spinal cord injury; shRNA: short hairpin RNA; Tyr-tubulin: tyrosinated-tubulin.
    Anti Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/anti gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    anti gfp - by Bioz Stars, 2023-09
    86/100 stars
      Buy from Supplier

    86
    Cell Signaling Technology Inc gfp
    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing <t>luciferase-GFP</t> were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.
    Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    gfp - by Bioz Stars, 2023-09
    86/100 stars
      Buy from Supplier

    86
    Cell Signaling Technology Inc anti gfp antibody
    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing <t>luciferase-GFP</t> were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.
    Anti Gfp Antibody, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/anti gfp antibody/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    anti gfp antibody - by Bioz Stars, 2023-09
    86/100 stars
      Buy from Supplier

    86
    Cell Signaling Technology Inc chicken anti gfp
    (A) Accessibility profiles of astrocyte-specific enhancers in the human whole-body accessibility atlas . Single-cell profiles were grouped within each tissue into pseudo-bulk aggregates, then normalized according to the signal (reads in peaks) within the dataset. Accessibility profiles are likely to predict enhancer activities within each tissue. Focusing on liver, some astrocyte-specific enhancers are predicted to have high expression, and some are predicted to have very little or no expression. In contrast, accessibility atlases do not predict expression of GFAP promoter across tissues. (B) Whole livers from mice injected intravenously with eHGT_381h- and eHGT_390m-enhancer-AAV vectors, stained <t>with</t> <t>anti-GFP</t> antibody. eHGT_381h has high liver accessibility, is predicted to have high liver expression, and shows many strong SYFP2-expressing hepatocytes throughout the liver as predicted. In contrast, eHGT_390m has very little liver accessibility and so is predicted to have little liver expression, and in fact shows few positive SYFP2-expressing hepatocytes as predicted. (C) Agreement between liver expression predictions and liver expression measurements across several astrocyte-specific enhancer-AAV vectors. eHGT_371m, 371h, 381h, and 386m all show many SYFP2-expressing hepatocytes as predicted. eHGT_390m, 390h, 375m, and 387m show few weak SYFP2-expressing hepatocytes as predicted. GFAP promoter shows many expressing hepatocytes, which was not predictable from the accessibility atlases. eHGT_380h shows many SYFP2-expressing astrocytes, in contrast to the epigenetic prediction. Liver images in B and C represent one to two mice analyzed for each vector. (D-E) Testing fidelity of enhancer-AAV expression across disease states. We used a Dravet syndrome model Scn1a R613X/+ mouse to induce epilepsy-associated hippocampal gliosis, injected enhancer-AAVs prior to the critical period, and analyzed tissue for expression patterns after the critical period (D). We assessed hippocampal gliosis with anti-GFAP antibody and enhancer-AAV expression <t>with</t> <t>anti-GFP</t> antibody (E). eHGT_390m maintained specific expression and similar levels in hippocampal astrocytes regardless of epileptic gliosis. In contrast, GFAP promoter expression strongly increased in gliotic astrocytes, and also was observed in dentate gyrus granule cells. Red dashed rectangles indicate the position of the expanded zoomed view, and the curved arrows indicate a rotated view. Abbreviations: ML molecular layer, GCL granule cell layer, PoL polymorphic layer.
    Chicken Anti Gfp, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/chicken anti gfp/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    chicken anti gfp - by Bioz Stars, 2023-09
    86/100 stars
      Buy from Supplier

    86
    Cell Signaling Technology Inc rabbit anti gfp antibody
    A. Left: Consensus motif of KEAP1 KELCH as established with the HD2 library and the aligned SQSTM1 peptide with the P348L mutation site indicated in blue. Right: Displacement curves from KEAP1 KELCH domain with wild-type/P348L SQSTM1 334-349 peptide. Measurements were in at least technical triplets. B. <t>GFP</t> trap of YFP or GFPKEAP1 and probing for co-immunoprecipitation of wild-type/P348L Myc-SQSTM1 in HeLa cells (n=3). C-D . Network of KEAP1 ( C ) or KPNA4 ( D ) with the prey proteins from the domain-mutation pairs. Interactions and corresponding mutations disrupted by the mutation are indicated in blue, enhanced in red and neutral effects in grey. A green dot indicates previously reported interacting prey proteins and a straight line if the domain’s binding motif is found in the binding peptide. The circle size encodes the confidence level of the domain-mutation pairs. For KPNA4 the network has been reduced to include the reported interactors and the domain-mutation pairs with an associated p-value ≤ 0.001. E. Left: Consensus motif of KPNA4 ARM as established with the HD2 library and the aligned CDC45 and ABRAXAS1 peptides with the mutation sites indicated in blue. Right: Displacement curves from KPNA4 ARM domain with wild-type/R157C CDC45 152-166 and wild-type/R361Q ABRAXAS1 349-364 peptides. Measurements were in technical triplets. F-G. Representative images of the localisation of wild-type and mutant EGFP-tagged (R361Q) ABRAXAS1 and (R157C) CDC45 in relation to the nuclear Hoechst staining (n=3). H. GFP trap of EGFP (n=3), wildtype EGFP-CDC45 (n=3) or mutant R157C EGFP-CDC45 (n=2) and probing for co-immunoprecipitation <t>of</t> <t>T7-KPNA7</t> in HEK293T cells.
    Rabbit Anti Gfp Antibody, supplied by Cell Signaling Technology Inc, used in various techniques. Bioz Stars score: 86/100, based on 1 PubMed citations. ZERO BIAS - scores, article reviews, protocol conditions and more
    https://www.bioz.com/result/rabbit anti gfp antibody/product/Cell Signaling Technology Inc
    Average 86 stars, based on 1 article reviews
    Price from $9.99 to $1999.99
    rabbit anti gfp antibody - by Bioz Stars, 2023-09
    86/100 stars
      Buy from Supplier

    Image Search Results


    Depletion of Fign improves functional recovery by increasing Tyr-tubulin. (A, B) Western blotting of tyrosinated (Tyr)-tubulin and Fign after spinal cord contusion (A) and sciatic nerve crush (B). α/β-Tubulin was used as a loading control. The data are shown as mean ± SE ( n = 3). The data were normalized by control (0 day) group. Linear regression showed expression trends of target proteins after injury. (C, D) Model of Fign depletion in spinal cord injury: U6-GFP Fign-AAV9 virus injected at T8, spinal cord contusion at T10 (C). Timeline of virus injection, spinal cord contusion, and Basso-Beattie-Bresnahan (BBB) evaluation (D). (E) BBB scores after spinal cord contusion (mean ± SE, n = 12 or 13). * P < 0.05; # P < 0.05, vs . Ctrl shRNA group (two-way analysis of variance followed by post hoc Bonferroni’s test). (F) Representative western blotting of Fign from injured spinal cord at 14 days after spinal cord contusion. α/β-Tubulin was used as a loading control (mean ± SE, n = 4). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. (G) Representative immunostaining results of NF200 (red, Cy3) in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Immunopositivity of NF200 was markedly increased in the injured area in the Fign shRNA group. Scale bars: 500 μm, 100 μm (enlarged images). (H) Statistical results of relative NF200 levels (mean ± SE, n = 8, Student’s t -test). (I) Representative immunostaining results of Tyr-tubulin and EB3 in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Microtubule plus-end binding protein EB3 significantly increased and mostly concentrated in Tyr-tubulin enriched areas in the Fign shRNA group. Tyr-tubulin is labeled in red (Alexa Fluor 647) and EB3 in yellow (Cy3) (changed to green by pseudo-color). Scale bars: 500 μm, 100 μm (enlarged images). (J) Statistical results of relative Tyr-tubulin and EB3 levels (mean ± SE, n = 5). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. AAV: Adeno-associated virus; BBB: Basso-Beattie-Bresnahan; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; NF200: neurofilament-200; SCI: spinal cord injury; shRNA: short hairpin RNA; Tyr-tubulin: tyrosinated-tubulin.

    Journal: Neural Regeneration Research

    Article Title: Fidgetin interacting with microtubule end binding protein EB3 affects axonal regrowth in spinal cord injury

    doi: 10.4103/1673-5374.373716

    Figure Lengend Snippet: Depletion of Fign improves functional recovery by increasing Tyr-tubulin. (A, B) Western blotting of tyrosinated (Tyr)-tubulin and Fign after spinal cord contusion (A) and sciatic nerve crush (B). α/β-Tubulin was used as a loading control. The data are shown as mean ± SE ( n = 3). The data were normalized by control (0 day) group. Linear regression showed expression trends of target proteins after injury. (C, D) Model of Fign depletion in spinal cord injury: U6-GFP Fign-AAV9 virus injected at T8, spinal cord contusion at T10 (C). Timeline of virus injection, spinal cord contusion, and Basso-Beattie-Bresnahan (BBB) evaluation (D). (E) BBB scores after spinal cord contusion (mean ± SE, n = 12 or 13). * P < 0.05; # P < 0.05, vs . Ctrl shRNA group (two-way analysis of variance followed by post hoc Bonferroni’s test). (F) Representative western blotting of Fign from injured spinal cord at 14 days after spinal cord contusion. α/β-Tubulin was used as a loading control (mean ± SE, n = 4). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. (G) Representative immunostaining results of NF200 (red, Cy3) in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Immunopositivity of NF200 was markedly increased in the injured area in the Fign shRNA group. Scale bars: 500 μm, 100 μm (enlarged images). (H) Statistical results of relative NF200 levels (mean ± SE, n = 8, Student’s t -test). (I) Representative immunostaining results of Tyr-tubulin and EB3 in the spinal cord lesion zone at 14 days after spinal cord contusion and treatment with Ctrl or Fign shRNA. Microtubule plus-end binding protein EB3 significantly increased and mostly concentrated in Tyr-tubulin enriched areas in the Fign shRNA group. Tyr-tubulin is labeled in red (Alexa Fluor 647) and EB3 in yellow (Cy3) (changed to green by pseudo-color). Scale bars: 500 μm, 100 μm (enlarged images). (J) Statistical results of relative Tyr-tubulin and EB3 levels (mean ± SE, n = 5). The data were normalized by Ctrl shRNA group. The data were analyzed by Student’s t -test. AAV: Adeno-associated virus; BBB: Basso-Beattie-Bresnahan; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; NF200: neurofilament-200; SCI: spinal cord injury; shRNA: short hairpin RNA; Tyr-tubulin: tyrosinated-tubulin.

    Article Snippet: After transfer to a polyvinylidene fluoride membrane (Millipore, Boston, MA, USA), the membrane was blocked and then incubated overnight with primary antibodies at 4°C: anti-α/β tubulin (rabbit, 1:2000, CST, Danvers, MA, USA, Cat# 2148, RRID: AB_2288042), anti-tyrosinated tubulin (rat, 1:800, Abcam, Cambridge, UK, Cat# ab6160, RRID: AB_305328), anti-acetylated tubulin (mouse, 1:800, Abcam, Cat# ab24610, RRID: AB_448182), anti-Fign (mouse, 1:400, Santa Cruz Biotechnology, Santa Cruz, CA, USA, Cat# sc-514956), anti-GFP (rabbit, 1:1000, CST, Cat# 2555, RRID: AB_10692764), and anti-EB3 (rabbit, 1:20,000, Abcam, Cat# ab157217, RRID: AB_2890656).

    Techniques: Functional Assay, Western Blot, Expressing, Injection, shRNA, Immunostaining, Binding Assay, Labeling

    Fign regulates Tyr-tubulin by binding to EB3 and parking at microtubules plus-ends. (A) Representative inverted immunostaining of Fign (green, Alexa Fluor 488), tyrosinated (Tyr)-tubulin (red, Cy3), and actin (violet, Alexa Fluor 647). Scale bars: 20 μm, 5 μm (enlarged images). (B) Representative western blotting with GFP antibody after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. (C) Representative western blotting of tubulin proteins after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. The data (normalized by control GFP plasmid transfection) are shown as mean ± SE ( n = 3), and were analyzed by Student’s t -test. (D) Representative inverted immunostaining of Tyr-tubulin (red, Cy3) and acetylated-tubulin (green, Alexa Fluor 488) after E18 cortical neurons were transfected with Ctrl or EB3 siRNA and lentivirus (LV)5-NC or LV5-Fign. Ctrl siRNA combined with LV5-Fign decreased Tyr-tubulin and axonal length. EB3 siRNA combined with LV5-Fign rescued this phenomenon. Scale bar: 20 μm. (E) Statistical results of relative Tyr-tubulin levels (left panel, n = 15, normalized by Ctrl siRNA + LV5-NC group) and axonal length (right panel, n = 35) of neurons transfected with Ctrl or EB3 siRNA, together with LV5-NC or LV5-Fign. The data are shown as mean ± SE and were analyzed using one-way analysis of variance and Dunnett’s test. (F) Representative inverted immunostaining of Fign in E18 cortical neurons transfected with LV5-NC or LV5-Fign. Scale bar: 20 μm. Red dotted box shows Fign in the axon. Red arrow shows the axonal terminal. Ace-tubulin: Acetylated-tubulin; E18: rat embryo at 18 days; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; LV: lentivirus; P: pellet, tubulins in polymerized microtubules; S: supernatant, soluble tubulins; siRNA: small interfering RNA; T: total, total tubulins; Tyr-tubulin: tyrosinated-tubulin.

    Journal: Neural Regeneration Research

    Article Title: Fidgetin interacting with microtubule end binding protein EB3 affects axonal regrowth in spinal cord injury

    doi: 10.4103/1673-5374.373716

    Figure Lengend Snippet: Fign regulates Tyr-tubulin by binding to EB3 and parking at microtubules plus-ends. (A) Representative inverted immunostaining of Fign (green, Alexa Fluor 488), tyrosinated (Tyr)-tubulin (red, Cy3), and actin (violet, Alexa Fluor 647). Scale bars: 20 μm, 5 μm (enlarged images). (B) Representative western blotting with GFP antibody after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. (C) Representative western blotting of tubulin proteins after transfection of GFP or GFP-Fign plasmid to Neuro-2a cells for 2 days. The data (normalized by control GFP plasmid transfection) are shown as mean ± SE ( n = 3), and were analyzed by Student’s t -test. (D) Representative inverted immunostaining of Tyr-tubulin (red, Cy3) and acetylated-tubulin (green, Alexa Fluor 488) after E18 cortical neurons were transfected with Ctrl or EB3 siRNA and lentivirus (LV)5-NC or LV5-Fign. Ctrl siRNA combined with LV5-Fign decreased Tyr-tubulin and axonal length. EB3 siRNA combined with LV5-Fign rescued this phenomenon. Scale bar: 20 μm. (E) Statistical results of relative Tyr-tubulin levels (left panel, n = 15, normalized by Ctrl siRNA + LV5-NC group) and axonal length (right panel, n = 35) of neurons transfected with Ctrl or EB3 siRNA, together with LV5-NC or LV5-Fign. The data are shown as mean ± SE and were analyzed using one-way analysis of variance and Dunnett’s test. (F) Representative inverted immunostaining of Fign in E18 cortical neurons transfected with LV5-NC or LV5-Fign. Scale bar: 20 μm. Red dotted box shows Fign in the axon. Red arrow shows the axonal terminal. Ace-tubulin: Acetylated-tubulin; E18: rat embryo at 18 days; EB3: end binding protein 3; Fign: fidgetin; GFP: green fluorescent protein; LV: lentivirus; P: pellet, tubulins in polymerized microtubules; S: supernatant, soluble tubulins; siRNA: small interfering RNA; T: total, total tubulins; Tyr-tubulin: tyrosinated-tubulin.

    Article Snippet: After transfer to a polyvinylidene fluoride membrane (Millipore, Boston, MA, USA), the membrane was blocked and then incubated overnight with primary antibodies at 4°C: anti-α/β tubulin (rabbit, 1:2000, CST, Danvers, MA, USA, Cat# 2148, RRID: AB_2288042), anti-tyrosinated tubulin (rat, 1:800, Abcam, Cambridge, UK, Cat# ab6160, RRID: AB_305328), anti-acetylated tubulin (mouse, 1:800, Abcam, Cat# ab24610, RRID: AB_448182), anti-Fign (mouse, 1:400, Santa Cruz Biotechnology, Santa Cruz, CA, USA, Cat# sc-514956), anti-GFP (rabbit, 1:1000, CST, Cat# 2555, RRID: AB_10692764), and anti-EB3 (rabbit, 1:20,000, Abcam, Cat# ab157217, RRID: AB_2890656).

    Techniques: Binding Assay, Immunostaining, Western Blot, Transfection, Plasmid Preparation, Small Interfering RNA

    a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.

    Journal: Nature cancer

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    doi: 10.1038/s43018-023-00553-8

    Figure Lengend Snippet: a-b, KPC1 PDAC (a) or KP1 LUAD (b) tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg body weight) and palbociclib (100 mg/kg body weight) (T/P) for 2 weeks (left). Right, flow cytometry analysis of NK cell numbers and degranulation in each condition (a) NK cell numbers PIP V, PIP TP and PIL TP, n=10; PIL V, n=9) and degranulation (n=5 independent mice per group). (b) NK cell numbers LIL V, n=3: LIL TP, n=5; LIP V, n=13; LIP TP, n=15 and degranulation LIP V, n=5; LIP TP, n=7 independent mice). Data represents pool of 3 independent experiments. c, KPC1 PDAC or KP1 LUAD cells expressing luciferase-GFP were injected orthotopically into the livers of 8-12 week old C57BL/6 female mice and treated as in (a) following tumor formation (left). Right, flow cytometry analysis of NK cell numbers Data represents pool of 2 independent experiments. (c) NK cell numbers and NK degranulation PILiver V, n= 9; PILiver TP, n=10 and LILiver V, n=8; LILiver TP, n=10 independent mice). d, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in pancreas (PIP) treated with vehicle or trametinib (1 mg/kg) and palbociclib (100 mg/kg) in the presence or absence of a NK1.1 depleting antibody (PK136; 250 ug) (V, n=5; TP and TP+αNK1.1, n=7 independent mice). e, Kaplan-Meier survival curve of C57BL/6 mice harboring KPC1 PDAC tumors in lungs (PIL) and treated as in (d) (V, n=10; TP, n=9 and TP+αNK1.1, n=8 independent mice). f, Kaplan-Meier survival curve of C57BL/6 mice harboring KP2 LUAD tumors in the lungs (LIL) and treated as in (d) (V, n=6; TP, n=7 and TP+αNK1.1, n=8 independent mice). g, Kaplan-Meier survival curve of C57BL/6 mice harboring KP1 LUAD tumors in pancreas (LIP) and treated as in (d) (V, n=7; TP and TP+αNK1.1, n=8 independent mice). P values in a-c were calculated using two-tailed, unpaired Student’s t-test, and those in d-g calculated using log-rank test. Error bars, mean ± SEM.

    Article Snippet: The following primary antibodies were used: H3K27me3 (9733; 1:400) and GFP (2956; 1:200) (Cell Signaling); p21 (556431; 1:200) (BD Biosciences); and GFP (AB6673; 1:250) (Abcam).

    Techniques: Expressing, Luciferase, Injection, Flow Cytometry, Two Tailed Test

    a, Representative Haematoxylin and eosin (H&E) (top) and Masson’s trichrome (bottom) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors from 2-3 independent experiments. Scale bars, 100μm. b, Immunohistochemical (IHC) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage of SA-β-gal+ area and the number of Ki67+ and pRb+ cells per field are shown inset (n=2-4 per group). Scale bar, 50μm. c, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors grown in different organs and treated as in (b). Quantification of the percentage of GFP+ (green) tumor cells expressing p21 (cyan) is shown inset (n=2-4 per group). Scale bar, 50μm. d, GFP+ tumor cells were FACS sorted from indicated tumors and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). Gene Set Enrichment Analysis (GSEA) of RNA-seq data using an established senescence gene set is shown. NES, normalized enrichment score. P values in d were calculated using two-sided, Kolmogorov-Smirnov test. Error bars, mean ± SEM.

    Journal: Nature cancer

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    doi: 10.1038/s43018-023-00553-8

    Figure Lengend Snippet: a, Representative Haematoxylin and eosin (H&E) (top) and Masson’s trichrome (bottom) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors from 2-3 independent experiments. Scale bars, 100μm. b, Immunohistochemical (IHC) staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage of SA-β-gal+ area and the number of Ki67+ and pRb+ cells per field are shown inset (n=2-4 per group). Scale bar, 50μm. c, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD (LIL, LIP, LILiver) derived-tumors grown in different organs and treated as in (b). Quantification of the percentage of GFP+ (green) tumor cells expressing p21 (cyan) is shown inset (n=2-4 per group). Scale bar, 50μm. d, GFP+ tumor cells were FACS sorted from indicated tumors and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). Gene Set Enrichment Analysis (GSEA) of RNA-seq data using an established senescence gene set is shown. NES, normalized enrichment score. P values in d were calculated using two-sided, Kolmogorov-Smirnov test. Error bars, mean ± SEM.

    Article Snippet: The following primary antibodies were used: H3K27me3 (9733; 1:400) and GFP (2956; 1:200) (Cell Signaling); p21 (556431; 1:200) (BD Biosciences); and GFP (AB6673; 1:250) (Abcam).

    Techniques: In Vivo, Staining, Derivative Assay, Immunohistochemistry, Immunofluorescence, Expressing, RNA Sequencing Assay

    a, KPC1 PDAC or KP1 LUAD tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. GFP+ tumor cells were FACS sorted and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). b, KEGG pathway analysis of pathways enriched in tumors in the lungs (LIL, PIL) compared to tumors in the pancreas (PIP, LIP) following T/P treatment. c, Heatmap showing fold change in SASP gene expression following T/P treatment in indicated tumor settings. d, Fold change in expression of select SASP chemokines following T/P treatment in indicated tumor settings (n=2-4 per group). Error bars, mean ± SEM. e, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumors in the pancreas (PIP, LIP) following T/P treatment. f, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. g, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL) and KP1 LUAD (LIL, LIP) derived-tumors grown in different organs and treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (PIP V; PIP TP; PIL TP; LIL TP and LIP TP, n=3, PIL V; LIL V and LIP V, n=2 independent tumors). Scale bar, 50μm. P values in b were calculated using two-sided, hypergeometric test and those in f using two-sided, Kolmogorov-Smirnov test.

    Journal: Nature cancer

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    doi: 10.1038/s43018-023-00553-8

    Figure Lengend Snippet: a, KPC1 PDAC or KP1 LUAD tumor cells expressing luciferase-GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation in the lungs or pancreas, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. GFP+ tumor cells were FACS sorted and extracted RNA subjected to RNA-seq analysis (n=2-4 per group). b, KEGG pathway analysis of pathways enriched in tumors in the lungs (LIL, PIL) compared to tumors in the pancreas (PIP, LIP) following T/P treatment. c, Heatmap showing fold change in SASP gene expression following T/P treatment in indicated tumor settings. d, Fold change in expression of select SASP chemokines following T/P treatment in indicated tumor settings (n=2-4 per group). Error bars, mean ± SEM. e, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumors in the pancreas (PIP, LIP) following T/P treatment. f, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. g, Immunofluorescence staining of indicated KPC1 PDAC (PIP, PIL) and KP1 LUAD (LIL, LIP) derived-tumors grown in different organs and treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (PIP V; PIP TP; PIL TP; LIL TP and LIP TP, n=3, PIL V; LIL V and LIP V, n=2 independent tumors). Scale bar, 50μm. P values in b were calculated using two-sided, hypergeometric test and those in f using two-sided, Kolmogorov-Smirnov test.

    Article Snippet: The following primary antibodies were used: H3K27me3 (9733; 1:400) and GFP (2956; 1:200) (Cell Signaling); p21 (556431; 1:200) (BD Biosciences); and GFP (AB6673; 1:250) (Abcam).

    Techniques: Expressing, Luciferase, Injection, RNA Sequencing Assay, Immunofluorescence, Staining, Derivative Assay

    a, Schematic of KPC PDAC syngeneic orthotopic transplantation into 8-16 week old male and female SMA-TK mice and treatment regimens. b, Immunohistochemical (IHC) staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated with vehicle, trametinib (1 mg/kg) and palbociclib (100 mg/kg), and/or ganciclovir (GCV) (50 mg/kg) for 2 weeks. Quantification of number of SMA+ cells per field is shown inset (b, V, n=5; TP, n=4; GCV, n=4; and TP/GCV, n=5 independent tumors). Scale bar, 50μm. c, Waterfall plot of the response of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice to treatment as in (b) (c, V, n=6; TP, n=10; GCV, n=8, and TP/GCV, n=15 independent mice). Data represents pool of 2 independent experiments. d-e, Flow cytometry analysis of NK (d) and T cell (e) numbers and activation markers in KPC1 orthotopic PDAC tumors propagated in SMA-TK mice following treatment as in (b) (n=8-16 per group). (d-e, V, n=8; TP, n=9; GCV, n=8, and TP/GCV, n=16 independent mice). Data represents pool of 2 independent experiments. Error bars, mean ± SEM. f, KEGG (left) and REACTOME (right) pathway analysis of RNA-seq data generated from FACS sorted GFP+ tumor cells from SMA-TK mice harboring KPC1 orthotopic PDAC tumors and treated as in (b) (n=4-5 per group). g, Heatmap of RNA-seq analysis of SASP gene expression in PDAC cells from KPC1 orthotopic PDAC tumors propagated in SMA-TK mice and treated as in (b) (n=4-5 per group). h, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumor cells from KPC1 orthotopic PDAC propagated in SMA-TK mice treated with T/P alone compared with combined T/P/GCV treatment. i, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. j, Immunofluorescence staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated as in (b). Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (n=2 per group). Scale bar, 50μm. P values in c-e were calculated using two-tailed, unpaired Student’s t-test, f using two-sided, hypergeometric test, and i using two-sided, Kolmogorov-Smirnov test.

    Journal: Nature cancer

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    doi: 10.1038/s43018-023-00553-8

    Figure Lengend Snippet: a, Schematic of KPC PDAC syngeneic orthotopic transplantation into 8-16 week old male and female SMA-TK mice and treatment regimens. b, Immunohistochemical (IHC) staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated with vehicle, trametinib (1 mg/kg) and palbociclib (100 mg/kg), and/or ganciclovir (GCV) (50 mg/kg) for 2 weeks. Quantification of number of SMA+ cells per field is shown inset (b, V, n=5; TP, n=4; GCV, n=4; and TP/GCV, n=5 independent tumors). Scale bar, 50μm. c, Waterfall plot of the response of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice to treatment as in (b) (c, V, n=6; TP, n=10; GCV, n=8, and TP/GCV, n=15 independent mice). Data represents pool of 2 independent experiments. d-e, Flow cytometry analysis of NK (d) and T cell (e) numbers and activation markers in KPC1 orthotopic PDAC tumors propagated in SMA-TK mice following treatment as in (b) (n=8-16 per group). (d-e, V, n=8; TP, n=9; GCV, n=8, and TP/GCV, n=16 independent mice). Data represents pool of 2 independent experiments. Error bars, mean ± SEM. f, KEGG (left) and REACTOME (right) pathway analysis of RNA-seq data generated from FACS sorted GFP+ tumor cells from SMA-TK mice harboring KPC1 orthotopic PDAC tumors and treated as in (b) (n=4-5 per group). g, Heatmap of RNA-seq analysis of SASP gene expression in PDAC cells from KPC1 orthotopic PDAC tumors propagated in SMA-TK mice and treated as in (b) (n=4-5 per group). h, Transcription factor enrichment analysis showing transcriptional regulators whose targets are differentially expressed in tumor cells from KPC1 orthotopic PDAC propagated in SMA-TK mice treated with T/P alone compared with combined T/P/GCV treatment. i, Gene Set Enrichment Analysis (GSEA) of EZH2 transcriptional targets. NES, normalized enrichment score. j, Immunofluorescence staining of KPC1 orthotopic PDAC tumors propagated in SMA-TK mice treated as in (b). Quantification of the percentage GFP+ (green) tumor cells expressing H3K27me3 (cyan) is shown inset (n=2 per group). Scale bar, 50μm. P values in c-e were calculated using two-tailed, unpaired Student’s t-test, f using two-sided, hypergeometric test, and i using two-sided, Kolmogorov-Smirnov test.

    Article Snippet: The following primary antibodies were used: H3K27me3 (9733; 1:400) and GFP (2956; 1:200) (Cell Signaling); p21 (556431; 1:200) (BD Biosciences); and GFP (AB6673; 1:250) (Abcam).

    Techniques: Transplantation Assay, Immunohistochemistry, Flow Cytometry, Activation Assay, RNA Sequencing Assay, Generated, Expressing, Immunofluorescence, Staining, Two Tailed Test

    a-b, KPC2 PDAC or KP2 LUAD tumor cells expressing GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Flow cytometry analysis of NK cell numbers and degranulation in PDAC (PIP, PIL) (a) and LUAD-derived tumors (LIL, LIP) (b) grown in different organs are shown (a-b, PIP V, n=3; PIP TP, n=4; PIL V and PIL TP, n=8 independent mice). c, Flow cytometry analysis of NK cell numbers and degranulation in spleens of mice with KPC1-derived PIP tumors treated as in (a) (n=5 independent mice per group). d, Kaplan-Meier survival curve of mice with KPC2-derived PIP tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) (d, V, n=5; TP; TP+αNK1.1 and TP+αCD8, n=8). e, IVIS images showing luciferase signaling in KPC1-derived PIL tumors following treatment as in (a). Right, quantification of total luminescence in the thoracic region (e, V, n=5; TP and TP+αNK1.1, n=8 independent mice). f, Waterfall plot of the response of KPC1-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 depleting antibody (PK136; 250 μg) (f, V and TP, n=5, TP+αNK1.1, n=6 independent mice). g, Waterfall plot of the response of KPC2-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) depleting antibody (g, V, n=5; TP, n=7; TP+αNK1.1 and TP+αCD8 ,n=8 independent mice). h, Kaplan-Meier survival curve of mice with KPC1-derived PIL tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against CD8 (2.43; 200 μg) or CD4 (GK1.5; 200 μg) (h, V, n=5; TP, TP+αNK1.1 and TP+αCD8, n=7 independent mice). i, Flow cytometry analysis of CD4+ and CD8+ T cell numbers and degranulation in KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD-derived tumors (LIL, LIP, LILiver) grown in different organs and treated as in (a) (i, PIP V, PIP TP, PIL TP, n=10; PIL V, n=9; LIL V, n=3; LIL TP, n=5; LIP V, n=13; LIP TP, n=15; PILiver V, n=9; PILiver TP, n=10; LILiver V, n=8; LILiver TP, n=10 independent mice). Data represents pool of 3 independent experiments. P values in a-c, e-g, and i were calculated using two-tailed, unpaired Student’s t-test, and those in d and h were calculated using log-rank test. Error bars, mean ± SEM.

    Journal: Nature cancer

    Article Title: EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance

    doi: 10.1038/s43018-023-00553-8

    Figure Lengend Snippet: a-b, KPC2 PDAC or KP2 LUAD tumor cells expressing GFP were injected i.v. or orthotopically into the pancreas of 8-12 week old C57BL/6 female mice. Following tumor formation, mice were treated with vehicle (V) or combined trametinib (1mg/kg) and palbociclib (100 mg/kg) (T/P) for 2 weeks. Flow cytometry analysis of NK cell numbers and degranulation in PDAC (PIP, PIL) (a) and LUAD-derived tumors (LIL, LIP) (b) grown in different organs are shown (a-b, PIP V, n=3; PIP TP, n=4; PIL V and PIL TP, n=8 independent mice). c, Flow cytometry analysis of NK cell numbers and degranulation in spleens of mice with KPC1-derived PIP tumors treated as in (a) (n=5 independent mice per group). d, Kaplan-Meier survival curve of mice with KPC2-derived PIP tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) (d, V, n=5; TP; TP+αNK1.1 and TP+αCD8, n=8). e, IVIS images showing luciferase signaling in KPC1-derived PIL tumors following treatment as in (a). Right, quantification of total luminescence in the thoracic region (e, V, n=5; TP and TP+αNK1.1, n=8 independent mice). f, Waterfall plot of the response of KPC1-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 depleting antibody (PK136; 250 μg) (f, V and TP, n=5, TP+αNK1.1, n=6 independent mice). g, Waterfall plot of the response of KPC2-derived PIP tumors following 2 week treatment with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or an NK1.1 (PK136; 250 μg) or CD8 (2.43; 200 μg) depleting antibody (g, V, n=5; TP, n=7; TP+αNK1.1 and TP+αCD8 ,n=8 independent mice). h, Kaplan-Meier survival curve of mice with KPC1-derived PIL tumors treated with vehicle, combined trametinib (1mg/kg) and palbociclib (100mg/kg), and/or depleting antibodies against CD8 (2.43; 200 μg) or CD4 (GK1.5; 200 μg) (h, V, n=5; TP, TP+αNK1.1 and TP+αCD8, n=7 independent mice). i, Flow cytometry analysis of CD4+ and CD8+ T cell numbers and degranulation in KPC1 PDAC (PIP, PIL, PILiver) and KP1 LUAD-derived tumors (LIL, LIP, LILiver) grown in different organs and treated as in (a) (i, PIP V, PIP TP, PIL TP, n=10; PIL V, n=9; LIL V, n=3; LIL TP, n=5; LIP V, n=13; LIP TP, n=15; PILiver V, n=9; PILiver TP, n=10; LILiver V, n=8; LILiver TP, n=10 independent mice). Data represents pool of 3 independent experiments. P values in a-c, e-g, and i were calculated using two-tailed, unpaired Student’s t-test, and those in d and h were calculated using log-rank test. Error bars, mean ± SEM.

    Article Snippet: The following primary antibodies were used: H3K27me3 (9733; 1:400) and GFP (2956; 1:200) (Cell Signaling); p21 (556431; 1:200) (BD Biosciences); and GFP (AB6673; 1:250) (Abcam).

    Techniques: Expressing, Injection, Flow Cytometry, Derivative Assay, Luciferase, Two Tailed Test

    (A) Accessibility profiles of astrocyte-specific enhancers in the human whole-body accessibility atlas . Single-cell profiles were grouped within each tissue into pseudo-bulk aggregates, then normalized according to the signal (reads in peaks) within the dataset. Accessibility profiles are likely to predict enhancer activities within each tissue. Focusing on liver, some astrocyte-specific enhancers are predicted to have high expression, and some are predicted to have very little or no expression. In contrast, accessibility atlases do not predict expression of GFAP promoter across tissues. (B) Whole livers from mice injected intravenously with eHGT_381h- and eHGT_390m-enhancer-AAV vectors, stained with anti-GFP antibody. eHGT_381h has high liver accessibility, is predicted to have high liver expression, and shows many strong SYFP2-expressing hepatocytes throughout the liver as predicted. In contrast, eHGT_390m has very little liver accessibility and so is predicted to have little liver expression, and in fact shows few positive SYFP2-expressing hepatocytes as predicted. (C) Agreement between liver expression predictions and liver expression measurements across several astrocyte-specific enhancer-AAV vectors. eHGT_371m, 371h, 381h, and 386m all show many SYFP2-expressing hepatocytes as predicted. eHGT_390m, 390h, 375m, and 387m show few weak SYFP2-expressing hepatocytes as predicted. GFAP promoter shows many expressing hepatocytes, which was not predictable from the accessibility atlases. eHGT_380h shows many SYFP2-expressing astrocytes, in contrast to the epigenetic prediction. Liver images in B and C represent one to two mice analyzed for each vector. (D-E) Testing fidelity of enhancer-AAV expression across disease states. We used a Dravet syndrome model Scn1a R613X/+ mouse to induce epilepsy-associated hippocampal gliosis, injected enhancer-AAVs prior to the critical period, and analyzed tissue for expression patterns after the critical period (D). We assessed hippocampal gliosis with anti-GFAP antibody and enhancer-AAV expression with anti-GFP antibody (E). eHGT_390m maintained specific expression and similar levels in hippocampal astrocytes regardless of epileptic gliosis. In contrast, GFAP promoter expression strongly increased in gliotic astrocytes, and also was observed in dentate gyrus granule cells. Red dashed rectangles indicate the position of the expanded zoomed view, and the curved arrows indicate a rotated view. Abbreviations: ML molecular layer, GCL granule cell layer, PoL polymorphic layer.

    Journal: bioRxiv

    Article Title: Enhancer-AAVs allow genetic access to oligodendrocytes and diverse populations of astrocytes across species

    doi: 10.1101/2023.09.20.558718

    Figure Lengend Snippet: (A) Accessibility profiles of astrocyte-specific enhancers in the human whole-body accessibility atlas . Single-cell profiles were grouped within each tissue into pseudo-bulk aggregates, then normalized according to the signal (reads in peaks) within the dataset. Accessibility profiles are likely to predict enhancer activities within each tissue. Focusing on liver, some astrocyte-specific enhancers are predicted to have high expression, and some are predicted to have very little or no expression. In contrast, accessibility atlases do not predict expression of GFAP promoter across tissues. (B) Whole livers from mice injected intravenously with eHGT_381h- and eHGT_390m-enhancer-AAV vectors, stained with anti-GFP antibody. eHGT_381h has high liver accessibility, is predicted to have high liver expression, and shows many strong SYFP2-expressing hepatocytes throughout the liver as predicted. In contrast, eHGT_390m has very little liver accessibility and so is predicted to have little liver expression, and in fact shows few positive SYFP2-expressing hepatocytes as predicted. (C) Agreement between liver expression predictions and liver expression measurements across several astrocyte-specific enhancer-AAV vectors. eHGT_371m, 371h, 381h, and 386m all show many SYFP2-expressing hepatocytes as predicted. eHGT_390m, 390h, 375m, and 387m show few weak SYFP2-expressing hepatocytes as predicted. GFAP promoter shows many expressing hepatocytes, which was not predictable from the accessibility atlases. eHGT_380h shows many SYFP2-expressing astrocytes, in contrast to the epigenetic prediction. Liver images in B and C represent one to two mice analyzed for each vector. (D-E) Testing fidelity of enhancer-AAV expression across disease states. We used a Dravet syndrome model Scn1a R613X/+ mouse to induce epilepsy-associated hippocampal gliosis, injected enhancer-AAVs prior to the critical period, and analyzed tissue for expression patterns after the critical period (D). We assessed hippocampal gliosis with anti-GFAP antibody and enhancer-AAV expression with anti-GFP antibody (E). eHGT_390m maintained specific expression and similar levels in hippocampal astrocytes regardless of epileptic gliosis. In contrast, GFAP promoter expression strongly increased in gliotic astrocytes, and also was observed in dentate gyrus granule cells. Red dashed rectangles indicate the position of the expanded zoomed view, and the curved arrows indicate a rotated view. Abbreviations: ML molecular layer, GCL granule cell layer, PoL polymorphic layer.

    Article Snippet: For IHC we used the following antibodies: chicken anti-GFP (Aves # GFP-1010), rabbit anti-Sox9 (Cell Signaling clone D8G8H, # 82630S), mouse CC1 antibody (Abcam # ab16794), mouse anti-GFAP (Millipore Sigma clone G-A-5, # G3893), with 5% normal goat serum (Thermo Fisher Scientific # 31872) and 0.1% Triton X-100 (VWR 97062-208) for blocking and permeabilization, and appropriate Alexa Fluor-conjugated secondary antibodies for detection.

    Techniques: Expressing, Injection, Staining, Plasmid Preparation

    A. Left: Consensus motif of KEAP1 KELCH as established with the HD2 library and the aligned SQSTM1 peptide with the P348L mutation site indicated in blue. Right: Displacement curves from KEAP1 KELCH domain with wild-type/P348L SQSTM1 334-349 peptide. Measurements were in at least technical triplets. B. GFP trap of YFP or GFPKEAP1 and probing for co-immunoprecipitation of wild-type/P348L Myc-SQSTM1 in HeLa cells (n=3). C-D . Network of KEAP1 ( C ) or KPNA4 ( D ) with the prey proteins from the domain-mutation pairs. Interactions and corresponding mutations disrupted by the mutation are indicated in blue, enhanced in red and neutral effects in grey. A green dot indicates previously reported interacting prey proteins and a straight line if the domain’s binding motif is found in the binding peptide. The circle size encodes the confidence level of the domain-mutation pairs. For KPNA4 the network has been reduced to include the reported interactors and the domain-mutation pairs with an associated p-value ≤ 0.001. E. Left: Consensus motif of KPNA4 ARM as established with the HD2 library and the aligned CDC45 and ABRAXAS1 peptides with the mutation sites indicated in blue. Right: Displacement curves from KPNA4 ARM domain with wild-type/R157C CDC45 152-166 and wild-type/R361Q ABRAXAS1 349-364 peptides. Measurements were in technical triplets. F-G. Representative images of the localisation of wild-type and mutant EGFP-tagged (R361Q) ABRAXAS1 and (R157C) CDC45 in relation to the nuclear Hoechst staining (n=3). H. GFP trap of EGFP (n=3), wildtype EGFP-CDC45 (n=3) or mutant R157C EGFP-CDC45 (n=2) and probing for co-immunoprecipitation of T7-KPNA7 in HEK293T cells.

    Journal: bioRxiv

    Article Title: Proteome-scale characterisation of protein motif interactome rewiring by disease mutations

    doi: 10.1101/2023.09.18.558189

    Figure Lengend Snippet: A. Left: Consensus motif of KEAP1 KELCH as established with the HD2 library and the aligned SQSTM1 peptide with the P348L mutation site indicated in blue. Right: Displacement curves from KEAP1 KELCH domain with wild-type/P348L SQSTM1 334-349 peptide. Measurements were in at least technical triplets. B. GFP trap of YFP or GFPKEAP1 and probing for co-immunoprecipitation of wild-type/P348L Myc-SQSTM1 in HeLa cells (n=3). C-D . Network of KEAP1 ( C ) or KPNA4 ( D ) with the prey proteins from the domain-mutation pairs. Interactions and corresponding mutations disrupted by the mutation are indicated in blue, enhanced in red and neutral effects in grey. A green dot indicates previously reported interacting prey proteins and a straight line if the domain’s binding motif is found in the binding peptide. The circle size encodes the confidence level of the domain-mutation pairs. For KPNA4 the network has been reduced to include the reported interactors and the domain-mutation pairs with an associated p-value ≤ 0.001. E. Left: Consensus motif of KPNA4 ARM as established with the HD2 library and the aligned CDC45 and ABRAXAS1 peptides with the mutation sites indicated in blue. Right: Displacement curves from KPNA4 ARM domain with wild-type/R157C CDC45 152-166 and wild-type/R361Q ABRAXAS1 349-364 peptides. Measurements were in technical triplets. F-G. Representative images of the localisation of wild-type and mutant EGFP-tagged (R361Q) ABRAXAS1 and (R157C) CDC45 in relation to the nuclear Hoechst staining (n=3). H. GFP trap of EGFP (n=3), wildtype EGFP-CDC45 (n=3) or mutant R157C EGFP-CDC45 (n=2) and probing for co-immunoprecipitation of T7-KPNA7 in HEK293T cells.

    Article Snippet: For visualisation, mouse anti-Myc antibody (1:1000, Thermo Scientific), mouse anti-Flag antibody (1:5000, Sigma Aldrich), rabbit anti-T7 (1:1000, Cell signalling), rabbit anti-GFP antibody (1:5000 in house) and mouse anti-GFP (1:1000, Roche) antibody were used and the membranes incubated overnight at 4 °C with the antibodies in 2.5 % milk in 1xPBS with 0.1% Tween-20.

    Techniques: Mutagenesis, Immunoprecipitation, Binding Assay, Staining