dedup-isc-ft-v107-score
float64
0.3
1
uid
stringlengths
32
32
text
stringlengths
1
17.9k
paper_id
stringlengths
8
11
original_image_filename
stringlengths
7
69
0.54824
ae66ddad430b42718b72ad6e6db76577
Longitudinal associations between antidepressants and WBC count. Asterisks (*) indicate associations passing multiple testing correction (<9.09 × 10−4).
PMC9892923
cxp-2023-0009-01-4-528605_f2.jpg
0.403164
5ee02535c4384de4a855f80b5c428511
Longitudinal associations between (a) SSRI, (b) SNRI, (c) TCA, and (d) atypical use and complete blood count and creatinine laboratory values. Asterisks (*) indicate associations passing multiple testing correction (<9.09 × 10−4).
PMC9892923
cxp-2023-0009-01-4-528605_f3.jpg
0.462902
2af57ef7d60046af84c553ea4ad09261
Longitudinal associations between antidepressant use and WBC values stratified by antidepressant indications (a) depression, (b) anxiety, and (c) chronic pain. Asterisks (*) indicate associations passing multiple testing correction (<9.09 × 10−4).
PMC9892923
cxp-2023-0009-01-4-528605_f4.jpg
0.483054
42a443f50ecf46799810e7bcc7726f2a
Schematic diagram of the CO2 flooding apparatus.
PMC9893740
ao2c06805_0002.jpg
0.414332
3952f50e6a354c1baec2d47e21472508
Diagenetic minerals and several types of pores in the samples. (A) Linear contacts of particles and intergranular pores in sample FDK-3. (B) Kaolinite in sample FDK-2. (C) Chlorite in sample FDK-3. (D) Mosaic shape ferrocalcite in sample FDK-5. (E) Combine pore with irregular shape in sample FDK-2. (F) Intercrystalline pores of illite in sample FDK-4.
PMC9893740
ao2c06805_0003.jpg
0.509178
9309d7f1990947dfa6bde1ba322f52bd
T2 distribution of NMR in samples from Chang 7 Member. (A–H) Samples FDK1–FDK8, respectively.
PMC9893740
ao2c06805_0004.jpg
0.512413
1b55690e4df14fc78dfce1fef82da19c
Plots of log(N(r)) versus log(T2) for two stages. (A–H) Samples FDK1–FDK8, respectively.
PMC9893740
ao2c06805_0005.jpg
0.413823
0b25a7ea17464239a5659865715d7acb
Relationship between corrosion rate and ferrocalcite content.
PMC9893740
ao2c06805_0006.jpg
0.492161
38aee89cffdf43f68929476e062c7360
Comparisons of T2 distribution curves before and after CO2 flooding in the sample from Chang 7 Member. (A–H) Samples FDK1–FDK8, respectively.
PMC9893740
ao2c06805_0007.jpg
0.497694
304ca861c94b4f25bafb26d508b71896
Plots of log(N(r)) versus log(T2) after CO2 flooding. (A–H) Samples FDK1–FDK8, respectively.
PMC9893740
ao2c06805_0008.jpg
0.443748
be489436ce0a4e67bfeb54813492fdaa
Comparison of pore distribution in (A) sample FDK2 and (B) sample FDK6.
PMC9893740
ao2c06805_0009.jpg
0.421565
6d869d2b60cd43e591bb896d52fed0f6
Microscopic characteristics in different lithofacies samples. (A) Chlorite coats the particle in sample FDK3. (B) Kaolinite fill pores in sample FDK1. (C) Intercrystalline pores connect with surrounding pores in sample FDK6. (D) Ferrocalcite fills the pores in a large cluster of crystals in sample FDK8.
PMC9893740
ao2c06805_0010.jpg
0.469451
1b76d600dea34e159e37abb1cb39ef65
Mineral content before and after CO2 flooding in (A) CL lithofacies and (B) CA lithofacies.
PMC9893740
ao2c06805_0011.jpg
0.408146
56262c48b15e46cd9e0aa9774071b5c3
Schematic diagrams of mineral changes after CO2 flooding in CL and CA lithofacies. (A) Mineral distribution in CL lithofacies before CO2 flooding. (B) Newly formed minerals fill the pores after CO2 flooding. (C) Ferrocalcite fills the pores in CA lithofacies before CO2 flooding. (D) Dissolution of ferrocalcite and new flow paths after CO2 flooding.
PMC9893740
ao2c06805_0012.jpg
0.492741
db2704ebed2547558fae27dec5b66435
Best overall response (A), PFS (B), OS (C), MRD (D), and blood cell immunophenotyping results at baseline and day 1/cycle 3 (predose) (E). (A–D) Patients included in efficacy population (n = 15): Isa 10 mg/kg-VCd, n = 12; Isa 20 mg/kg-VCd, n = 3. All = all patients in the efficacy population. MRD was evaluated in patients with ≥VGPR by NGF and NGS methods at 10−5 and MRD negativity rate determined by combining both methods in the case of at least 1 method yielding negative results and the other method showing no positive result at the same time. (E) Patients included in baseline population: Isa 10 mg/kg–VCd, n = 13; Isa 20 mg/kg–VCd, n = 4 and in on-treatment population: Isa 10 mg/kg-VCd, n = 10; Isa 20 mg/kg-VCd, n = 2. Results expressed as percentage of leukocytes. C = cyclophosphamide; C3D1 = cycle 3 day 1; CR = complete response; d = dexamethasone; Isa = isatuximab; MRD = minimal residual disease; NGF = next-generation flow; NGS = next-generation sequencing; NK = natural killer; ORR = overall response rate; OS = overall survival; PFS = progression-free survival; PR = partial response; sCR = stringent CR; SD = standard deviation; T-reg = regulatory T cell; V = bortezomib; VGPR = very good partial response.
PMC9894356
hs9-7-e829-g001.jpg
0.437816
51a7454ba0b44a5cac5ecd1f24aeb506
Study 1: Summary of Path Analyses.Note. Unstandardized path coefficients are reported. Dashed lines represent paths that are not significant. Control variables were included in the model, but their coefficients are not reported here. The model without control variables provides substantively equivalent results and does not change the interpretation of the relationships between variables (see Supplemental Online Materials).*p < .05. **p < .01.
PMC9895284
10.1177_19485506221077858-fig1.jpg
0.448241
6d486faa5c26439686e73959b29d522d
Study 2: Summary of Path Analyses.Note.a 1 = High Gratitude, 0 = Low Gratitude. Unstandardized path coefficients are reported. Dashed lines represent nonsignificant paths. Control variables were included in the model, but their coefficients are not reported here. The model without control variables provides substantively equivalent results and does not change the interpretation of the relationships between variables (see Supplemental Online Materials).*p < .05. **p < .01.
PMC9895284
10.1177_19485506221077858-fig2.jpg
0.40868
553d88fb12ae43cfbd22cc4f777f7f9d
Molecular design and interaction between FTPA and perovskite.a Molecular structure design of FTPA, and schematic diagram of the possible phase evolution of the nucleation and crystallization of FA-based mixed anion perovskites (FA0.95MA0.05Pb(I0.95Br0.05)3) during the film-forming process with (w) or without (w/o) FTPA. In the control perovskite film, the complicated intermediate phases, MA2Pb3I8·2DMSO and δ-FAPbI3, caused two competition pathways of crystal-nucleation as shown in Eqs. (1) and (2), and finally resulted in a low crystal orientation with δ-phase and PbI2. In contrast, perovskite film with FTPA restrained the formation of the intermediate phases and formed a hydrogen-bonding polymer network in the perovskite films which induced stable and preferred orientation of α-FAPbI3. b A comparison of 1H NMR spectra of FAI, FTPA/FAI, and FTPA/FAI/ PbI2. The photos are the FAI solution and FTPA/FAI mixture. Theoretical calculation show that the hydrogen bonds F⋯HN and O⋯HN formed between FTPA and FAI have a strength of −29.37 kcal mol−1. c FTIR spectra of the FAI, FTPA/FAI, and FTPA/FAI/PbI2. d Pb 4 f XPS spectra of the perovskite films of control and with FTPA, respectively.
PMC9895431
41467_2023_36224_Fig1_HTML.jpg
0.393102
85c7608a1c4e4faeb30b7cfb4394704f
In-situ monitoring of intermediate phase, nucleation, and crystallization process of the control and FTPA based perovskite films during spin-coating and annealing procedures.a In-situ tracking of X-ray diffraction of the perovskite films during three processes: wet perovskite film without (w/o) anti-solvent during spin-coating, wet perovskite film with (w) anti-solvent during spin-coating, and perovskite films annealed at 100 °C for various times. α and δ symbols indicated α-phase and δ-phase perovskite. b Optical microscopy images of the wet perovskite films without antisolvent during the spin-coating process. MA2Pb3I8·2DMSO film was prepared by perovskite of MAI/PbI2 (1:1 mol%) and δ-FAPbI3 was prepared by perovskite of FAI/PbI2 (1:1 mol%). c In-situ UV absorption spectra during the spin-coating process. d In-situ GIWAXS patterns during spin-coating process. e In-situ UV absorption spectra during the initial annealing process at 100 °C. f GIWAXS patterns of perovskite films after annealing 1 h.
PMC9895431
41467_2023_36224_Fig2_HTML.jpg
0.457831
1864dbb5c4204f85addfa0a963445bdd
Morphology, carrier extraction and energy level behaviors of the control, B-FTPA and BS-FTPA perovskite films.B-FTPA is the perovskite film with FTPA in the bulk, and BS-FTPA is the film further spin-coated 2 mg mL−1 FTPA and annealed for in-situ polymerizing on the surface of the B-FTPA. a Top-view SEM images. b HR-TEM image clearly shows the polymerized FTPA surround the grain boundaries of perovskite. J-V curves of c the electron-only device (FTO/SnO2/perovskite/PCBM/Ag) and d the hole-only device (FTO/PEDOT PSS/perovskite/Spiro-OMeTAD/Au) measured by SCLC model. The J-V curves can be divided into three regions that are the Ohmic region, trap-filled limit region (TFL), and Child’s region. The electron and hole trap density (Nt) are shown in Fig. 3c, d, respectively. e Time-resolved PL spectra. f Valence-band region and photoemission cut off energy of the UPS spectra. g Energy level scheme of the devices. Conduction band minimum (EC), Valence band maximum (EV), and Fermi level (EF).
PMC9895431
41467_2023_36224_Fig3_HTML.jpg
0.43941
7d8afcf3cc0b4b25a948ed2ea409ceab
Photovoltaic characteristics and stability of PSCs based on control, B-FTPA and BS-FTPA.a A cross-sectional SEM images showed the device architectures: FTO/SnO2/perovskite/Spiro-OMeTAD/Au for control, FTO/SnO2/B-FPTA perovskite (with FTPA 43.13 mg mL−1 in the bulk)/FTPA (2 mg mL−1)/Spiro-OMeTAD/ Au for BS-FTPA; ToF-SIMS cross-section images presented the element distribution in BS-FTPA. C12H10N- signals attributed to the diphenylamine branches of FTPA and Spiro-OMeTAD; F- signals corresponding to the Li-TFSI in Spiro-OMeTAD solution, FTPA in bulk and surface of perovskite, and FTO; the PbI3- signals corresponding to the perovskite. b J-V curves of the champion PSCs. c IPCE spectra of the devices integrated over the AM 1.5 G (100 mW cm−2) solar spectrum. d Semilog plot of IPCE at the absorption onset for BS-FTPA based PSCs, measured using FTPS at Jsc. An Urbach energy (Eu) of 14.5 meV can be obtained from the red line, a sharp absorption edge. e Statistical device data based on 30 devices. f Device stability of unencapsulated devices under1-sun illumination at 23 ± 2 °C in a nitrogen atmosphere (ISOS-LC-1). g Device stability of unencapsulated devices held at 25 ± 5 °C and 50 ± 10% relative humidity (RH) (ISOS-D-1). h Contact angle of perovskite films, and photographs of perovskite films dipped in water, FTO/SnO2/perovskite for control and FTO/SnO2/B-FTPA/FTPA for BS-FTPA. i Device stability of unencapsulated devices under 65 ± 3 °C thermal aging (ISOS-T-1). All of the error bars in Fig. 4f, g and i represent the standard deviation for six devices.
PMC9895431
41467_2023_36224_Fig4_HTML.jpg
0.449018
3e42a8bd4e44428386ed275c7eabbb41
Liver structure and zonation. A Schematic diagram of hepatic lobule and zonation. B A summary of hepatic zonation and its main function. Adapted from Halpern K.B.et al. 2017
PMC9895469
13619_2022_152_Fig1_HTML.jpg
0.43474
4d49860cd11e46869d32fb34359c5cea
Significant host biomarker concentrations determined in colon tissue homogenates of C3H/HeOuJ mice from the EPC experiment. a Cyanocobalamin supplementation enhanced IL-12/23p40 and IL-17A cytokines of naïve mice, with a higher trend of IL-4 levels (P = 0.06) compared to control. bC. rodentium challenge led to a significant increase in IFNγ, IL-10, IL-17A, and GM-CSF in the cyanocobalamin-supplemented mice (n = 8; *P < 0.05, **P < 0.01, ***P < 0.001)
PMC9896722
40168_2023_1461_Fig10_HTML.jpg
0.514985
aba4229bf03641f093540ff2f0514e2e
Cyanocobalamin-induced dysbiosis increased the production of the p40 subunit in colon tissues of mice prior to pathogen challenge. Pre-infection levels of IL-12/23p40 in colon tissues were directly related to microbiota structure and independent of related mice genotypes. a Gene expression was significantly higher for IL12B, the p40 subunit coding gene, but not for the IL-12A (p35) gene (n = 8) in the EPC experiment. b The SURV experiment did not display the increase in IL-12/23p40 (n = 5–6), nor did c germ-free C57BL/6J mice (n = 4; *P < 0.05, **P < 0.01, ***P < 0.001). d Cyanocobalamin and methylcobalamin supplementation at 10 μg/ml and 40 μg/ml increased IL/23p40 protein levels in the colon of conventional C57BL/6J mice (n = 6; P < 0.05) compared to control
PMC9896722
40168_2023_1461_Fig11_HTML.jpg
0.541171
ac8cb108d6994de4b7096c0f46f4b96a
The impact of cyanocobalamin supplementation on survival and early onset of C. rodentium infection in C3H/HeOuJ mice. a Supplementing cyanocobalamin in drinking water increased cecal and colon levels of cobalamin by ~1000 times (n = 15; *P < 0.05, **P < 0.01). b Daily fecal enumeration of C. rodentium in the EPC experiment indicated increased colonization burden at day 2 postinfection (D2PI) and D3PI from cyanocobalamin supplementation (n = 8; #P < 0.10, * P < 0.05). c Enumeration of C. rodentium at D5PI in the ileum and cecum was more consistent with cyanocobalamin supplementation, but colonization levels were similar (n = 8). d Consistent with the more rapid colonization, the SURV experiment revealed an earlier onset of mortality, reducing mice survival over the first 10-day postinfection (n = 9; P < 0.05; Mantel-Cox test). e Colon pathology scores at D5PI in naïve and infected mice show that cyanocobalamin significantly increased mucosal and epithelial damage (*) compared to infected control mice (n = 8; P < 0.05; limit of detection (LOD))
PMC9896722
40168_2023_1461_Fig1_HTML.jpg
0.517086
48d9757f9e874801b5498be98c0ffe79
Microbiota analyses of naïve and infected (inf_) mice from the EPC experiment. Principal coordinate plots of the ileum, cecum, and colon microbiota are based on the weighted and unweighted UniFrac dissimilarity metric. Distinct clustering was determined by unweighted UniFrac in the cecum and colon (Additional file 1 Table S2). Cyanocobalamin supplementation significantly reduced alpha diversity (observed and PD) in the colon (n = 7–12; P < 0.05)
PMC9896722
40168_2023_1461_Fig2_HTML.jpg
0.453673
706c02e9ccdb46168f7604c7e8b14bb8
DESeq2 differential analyses of microbial communities in mice from the EPC experiment. Cyanocobalamin supplementation altered the Firmicutes population throughout the GI tract. The control group had a greater abundance of Firmicutes, including Lachnospiraceae species and Clostridia vadinBB60 group bacterium in the cecum and colon a preinfection and b postinfection (n = 6–8; only ASVs with a P-value less than 0.05 were plotted; adjusted P-value was used for significance; bolded taxa represent a trend (P < 0.10); *P < 0.05, **P < 0.01, *** P < 0.001)
PMC9896722
40168_2023_1461_Fig3_HTML.jpg
0.447111
b9bc2f06f5054b45b9693aab506cb0bb
Comparison of the colonic microbial communities in naïve C3H/HeOuJ mice from the SURV and EPC experiments. a Unweighted UniFrac PCoA plot and b weighted UniFrac comparison show a similar pattern in microbial community clustering (Additional file 1 Table S2). c The changes were associated with a consistent reduction in alpha diversity (Observed and PD) regardless of experiment (n = 10–12; P < 0.05). Differential expression determined by DEseq2 analysis also showed that the Firmicutes populations (Clostridia vadinBB60 group) were impacted by cyanocobalamin in both mice harboring d SURV and e EPC gut microbiota (n = 10–12; bolded taxa represent a trend (P < 0.10; *P < 0.05, **P < 0.01, ***P < 0.001)
PMC9896722
40168_2023_1461_Fig4_HTML.jpg
0.434154
8e6f18eefb0c406786444f35d18e4bac
Cecal microbiota analysis of the 16S rRNA gene identified by SAMSA2 metatranscriptome analysis from the EPC experiment. Distinct clustering of microbial communities in a unweighted but not b weighted UniFrac PCoA plots (Additional file 1 Table S2). c Alpha diversity as determine by Observed, Shannon, and PD metrics showed that diversity (PD only) increased postinfection for cyanocobalamin-supplemented mice compared to control (P < 0.05). Differential expression analysis using DEseq2 of microbial taxa confirmed that the Firmicutes populations were altered from cyanocobalamin supplementation in d naïve and e infected mice (n = 6–8; bolded taxa represent a trend (P < 0.10); ***P < 0.001)
PMC9896722
40168_2023_1461_Fig5_HTML.jpg
0.395811
9588eb9d45c44c9ea5b47e0718e05a82
DESeq2 differential expression analyses of cecal metatranscriptome from the EPC experiment. a Naïve and bC. rodentium-challenged mice (inf_) supplemented with cyanocobalamin displayed altered functional activities related to metabolism (citrate:sodium symporter) and motility (flagellin domain protein) and confirmed to be from the Lachnospiraceae family (n = 8; bolded taxa represent a trend (P < 0.10); *P < 0.05, **P < 0.01)
PMC9896722
40168_2023_1461_Fig6_HTML.jpg
0.3992
9517b0199e984877b4d09794cd2eac45
SEED subsystems level 3 DESeq2 analysis of the C3H/HeOuJ cecal metatranscriptome from the EPC experiment. Cyanocobalamin supplementation increased pathways related to gram-negative lipopolysaccharide assembly while decreasing genes related to the competence of gram-positive bacteria in a naïve mice but not in bC. rodentium-challenged mice (n = 8; bolded taxa represent a trend (P < 0.10); *P < 0.05)
PMC9896722
40168_2023_1461_Fig7_HTML.jpg
0.435506
04e0ab8dafd04e0295193c91721a83c3
DESeq2 differential expression analyses of Lachnospiraceae and Citrobacter-specific functional activity from the EPC experiment. Prior to infection, the functional activity of the Lachnospiraceae family (a) shows that cyanocobalamin treatment increased the expression of numerous genes, including fibronectin type 3 domain-containing protein and serine/threonine transport SstT. b After exposure to C. rodentium, the Lachnospiraceae family members of the inf_CNCbl40 group displayed distinct activities compared to inf_CON group. Citrobacter-specific gene expression (c) was more pronounced in the inf_CNCbl40 group than inf_CON with notable signals of increased virulence gene expression, while control mice had more family 31 glucosidase activity (n = 8; bolded taxa represent a trend (P < 0.10); *P < 0.05, **P < 0.01)
PMC9896722
40168_2023_1461_Fig8_HTML.jpg
0.465803
ab42e9e788154a09b595b922dbeaa03b
In vitro competition assay of C. rodentium and B. thetaiotaomicron under physiological relevant concentrations of cyanocobalamin. Cyanocobalamin alone was unable to directly alter C. rodentium growth or virulence. a Enumeration of C. rodentium and B. thetaiotaomicron grown in competition for 6 h anaerobically and b of C. rodentium grown alone. Expression of gene related to virulence (Ler, Tir, and EspA) did not differ between treatments when C. rodentium was grown in c competition or d alone at different concentrations of cyanocobalamin (n = 6; P < 0.05)
PMC9896722
40168_2023_1461_Fig9_HTML.jpg
0.429182
5fd5f877deb4485e8ef6cf7cae42ee83
SEM images of Au cube (a), Au octahedron (b), Au tetradecahedron (c), Ag NPs (d), Au NRs (e) and Au NPs (f).
PMC9897048
d2ra07216e-f1.jpg
0.398234
d775797c3a4548d8bd8714e913348897
(a)Reproducibility characterization of gold nanorod substrates for detection of 10−11 M CV; (b). relative standard deviation (RSD) at the 1619 cm−1 characteristic peak of the CV molecule.
PMC9897048
d2ra07216e-f2.jpg
0.436226
df9e7320bb48440197b43e5b545c1cb5
Principle causing the solvent to move toward the smaller gaps of the nanocapillary pumping model. In the schematic diagram of Au NRs films, 1,2 and 3 represent small gaps between nanorods. Partial enlargement of the gap between two nanorods (b) and multiple nanorods (c). r1 and r2 represent the radius of curvature at different interstices, and P1 and P2 represent the pressures generated by the liquid at different interstices.
PMC9897048
d2ra07216e-f3.jpg
0.522775
2b991b9276874263b1c403b0c3ca97f2
SERS spectra at different concentrations. (a) GTX with concentrations ranging from 10−6 to 10−9 M. (b) NOD with concentrations ranging from 6 × 10−5 to 6 × 10−7 M. A plot of SERS intensity versus logarithmic (c) GTX, (d) NOD concentration for the band at 1609 and 1001 cm−1, respectively.
PMC9897048
d2ra07216e-f4.jpg
0.445172
621f15eb481f44b59ff375aab0eeb228
Psychosocial risk-factor screening questions and required response flow to qualify for the study.
PMC9897276
joem-65-e93-g001.jpg
0.592445
531ecd719705450aa11fac50e1b0cbad
Participant flow, from screening to onboarding on the Wysa app.
PMC9897276
joem-65-e93-g002.jpg
0.429141
4aee0a383910485486f3f241b5e3c371
The total number of sessions completed by all users across tools on the app.
PMC9897276
joem-65-e93-g003.jpg
0.432803
76c974e195b34341b7eb1bd876bebf99
Strategies to reduce regulatory cells in cancer patients. Several strategies alone or combining the targeting of different biochemical pathways have been reported to modulate regulatory cells in cancer patients. Some approaches aim to reduce the frequency, function and extent of blocking mobilization, while others focus on enabling phagocytosis, polarization, or potentiating differentiation. Blue crosses indicate potentiation/increase; forbidden sign indicates blockade/reduction
PMC9898962
12943_2023_1714_Fig1_HTML.jpg
0.484198
b563df0712f74a17ba16a60287a428f9
Effect of immunotherapy on Tregs. Several ICIs and other treatments can directly affect the number and function of regulatory T cells consequently, improving anti-tumoral function and preventing tumor growth. Some treatments act by reducing the number of Tregs (by antibody dependent cellular citotoxicity, ADCC) while others affect their suppressive activity or positively modulate the functions of CD8 T, NK cells and dendritic cells. The modulation towards pro-inflammatory cytokines and the increase in the CD4 + effector/Treg ratio are other mechanisms by which immunotherapy can potentiate the immune response within the tumor microenvironment. Created with Biorender.com
PMC9898962
12943_2023_1714_Fig2_HTML.jpg
0.49133
cf82c0c5618e49e8869c36b1e8da221b
Effect of immunotherapy on MDSCs. The figure depicts different treatments that can modulate MDSC expansion and function. The drugs affect the frequency of MDSCs, reduce MDSC suppressive activity, improve CD8 T cell function, and modulate the release of cytokines from the tumor. A combination of these therapies may have a synergistic effect that promotes better outcomes. Created with Biorender.com
PMC9898962
12943_2023_1714_Fig3_HTML.jpg
0.505114
460689512a724b44a978f3097550f14b
Effect of immunotherapy on TAMs. The figure displays treatments that aim to prevent the expansion of TAMs, consequently enhancing the anti-tumoral effect. The drugs work by promoting apoptosis (which reduces TAM frequencies)and TAM differentiation to M1 macrophages, and by blocking TAM mobilization, infiltration and metastasis. On the other hand, there is an increase in pro-inflammatory cytokine release and a reduction in the phagocytic activity of macrophages. Also, the use of these treatments combined with chemotherapy or other ICIs may be synergistic, bursting the immune response against the tumor. Created with Biorender.com
PMC9898962
12943_2023_1714_Fig4_HTML.jpg
0.509718
f3ae83c274054b3fbdf6f5216c4fc3bd
Lunasin inhibited the viability of breast cancer cells. (a) MCF-10A cells, (b) MDA-MB-231 cells, and (c) MCF-7 cells were treated with various doses of lunasin for 24, 48, and 72 h, and the cell viability was determined by MTT assay. The values of the data are presented as the mean ± SEM from at least three independent experiments. Statistical analysis was performed by independent sample t-test; significant differences are displayed as follows: *P < 0.05 for 24 h; #P < 0.05 for 48 h; †P < 0.05 for 72 h, which indicates lunasin treatment compared to the control group.
PMC9899045
FNR-67-8991-g001.jpg
0.416982
72a456bd2fba4fac9027401ea69e1ddf
The effects of lunasin on carcinogenic-related gene expression and protein secretion in breast cancer cells. Carcinogenic-related gene expression is shown in (a) MDA-MB-231 cells and (b) MCF-7 cells treated with lunasin for 24 h, and the gene expression was measured by qPCR. The specific gene expression is shown as the fold change compared with the control group. Cytokine production is shown in (c) MDA-MB-231 cells and (d) MCF-7 cells treated with lunasin for 24 and 48 h, and the supernatants were analyzed by ELISA. The value of data is presented as mean ± SEM, and statistical analysis was done by independent sample t-test; significant differences of control displayed in *P < 0.05.
PMC9899045
FNR-67-8991-g002.jpg
0.46284
af5af573c7fd4bcb878e44afe6c4bc86
The effects of lunasin on estrogen receptor and aromatase gene expression and aromatase activity in breast cancer cells. Gene expression is shown in (a) MDA-MB-231 cells and (b) MCF-7 cells treated with lunasin for 24 h, and the gene expression was measured by qPCR. The specific gene expression is shown as the fold change compared with the control group. The aromatase activity was determined in (c) MDA-MB-231 cells and (d) MCF-7 cells treated with lunasin for 48 h. The value of data is presented as mean ± SEM, and statistical analysis was done by independent sample t-test; significant differences of control displayed in *P < 0.05.
PMC9899045
FNR-67-8991-g003.jpg
0.452253
3455e51777fd417c844dc2b8c4601c33
The effects of β-estradiol and lunasin on cell viability in breast cancer cells. (a) MCF-10A cells, (b) MDA-MB-231 cells, and (c) MCF-7 cells were treated with various doses of β-estradiol (E2) for 24 and 48 h. Lunasin affected the viability of (d) MCF-10A cells, (e) MDA-MB-231 cells, and (f) MCF-7 cells treated with 20 nM E2 for 24 and 48 h, and the cell viability was determined by the MTT assay. The data are presented as the mean ± SEM from three independent experiments. Statistical analysis was performed by independent sample t-test; significant difference displayed in symbols: *P < 0.05, lunasin treatment versus control without E2; #P < 0.05, lunasin treatment versus control with E2.
PMC9899045
FNR-67-8991-g004.jpg
0.463361
4ff942dc2b824c99b94686c1331c41f6
Cell vitality of breast cancer cells treated with lunasin. MDA-MB-231 and MCF-7 cells were treated with lunasin for 48 h, MCF-7 cells were treated with or without 20 nM E2 at the same time, and the cells were collected. The cell vitality was analyzed: (a) definition of histogram plots and (b) fluorescent intensity plots, (c) the proportion of MDA-MB-231 cells, and (d) the proportion of MCF-7 cells. This figure represents the analytic pattern by the NC-3000, and the experiment was performed in at least three independent experiments.
PMC9899045
FNR-67-8991-g005.jpg
0.407146
e3a5b63e04084be7bb5e67d3b2421b6a
Cell apoptosis of breast cancer cells treated with lunasin. MDA-MB-231 and MCF-7 cells were treated with lunasin for 48 h, MCF-7 cells were treated with or without 20 nM E2 at the same time, and the cells were collected. Cell apoptosis was analyzed: (a) the definition of histogram plots and (b) fluorescent intensity plots, (c) the proportion of MDA-MB-231 cells, and (d) the proportion of MCF-7 cells. The figure represents the analytic pattern by the NC-3000, and the experiment was performed in at least three independent experiments.
PMC9899045
FNR-67-8991-g006.jpg
0.521636
110684eda89b4392a9da9c2b4d73c5ce
Proposed scheme of the possible mechanism by which lunasin suppresses the growth of breast cancer cells. Lunasin regulated inflammatory mediators, estrogen receptors gene expression, and aromatase activity, resulting in apoptosis induction and decreased cell viability and contributing to suppressing breast cancer progression. Abbreviations: COX-2, Cyclooxygenase-2; ER, estrogen receptor; IL, interleukin; PGE2, prostaglandin E2; VEGF, vascular endothelial growth factor.
PMC9899045
FNR-67-8991-g007.jpg
0.490719
8d28bbf28d3649e599673b9a8ee5bcc8
Comparison of linear regression models between Vcmax estimated from full A–Ci curves against apparent photosynthetic capacity estimated by the ‘one-point method’ (Vʹcmax; Equation 2) (A), and the modified version including the temperature dependency (Vʹcmax–ρ; Equation 5) (B). The light gray line is the 1:1 relationship.
PMC9899412
erac466f0001.jpg
0.378464
164c09ae09a240728eee3814634efb34
Residuals of maximum carboxylation rate, Vcmax, estimated from A–Ci curves estimated from apparent maximum carboxylation capacity, Vʹcmax (Equation 2) using the estimated Rday:Vcmax ratio (A), and using the temperature-dependent scaling factor (B) as a function of leaf temperature.
PMC9899412
erac466f0002.jpg
0.583958
1f0e3272c87b4f4baff7d2a047c4eb1a
The chalcogen-bonded chains in the crystal structure of ebselen 1.
PMC9899513
c-79-00043-fig1.jpg
0.535179
b941c4b8ee9e410ba6f2772bcbb4d27c
The hydrogen-bonding inter­actions in the structure of 2-Me+ tosyl­ate trihydrate. The undulating chain extends along the a axis.
PMC9899513
c-79-00043-fig10.jpg
0.583366
f552cd6b5c904aa0814cb4c273d11ac8
Hydrogen bonding, chalcogen bonding and π–π stacking in the structure of 2-Me+ tosyl­ate trihydrate.
PMC9899513
c-79-00043-fig11.jpg
0.496522
7c38349a53f8457baf6f7cb8e530dbd4
Experimentally determined electrostatic potential for com­pound 2 mapped onto the 0.05 a.u. isosurface.
PMC9899513
c-79-00043-fig12.jpg
0.525664
fda97c8c85f8462bb6d8f404549a3bd3
Critical points (CPs) in the vicinity of the Se atom for com­pound 2. (3,−1) CPs are shown in red (intra­molecular) and green (inter­molecular), and (3,+1) CPs are shown in blue.
PMC9899513
c-79-00043-fig13.jpg
0.393972
8e86f2a01d544fd49b19e0bf607219a5
Electron localization function (ELF) in the plane of the ring system for com­pound 2.
PMC9899513
c-79-00043-fig14.jpg
0.422926
05b7db2757c54675b492b1d83103078d
ELF plotted along the N1—Se1—N2 and C1—Se1—O1 bonds for com­pound 2. BCPs are shown as vertical dashed lines.
PMC9899513
c-79-00043-fig15.jpg
0.554532
97a88029c514484f91fcc8ffba11d80a
Difference electron residuals from the multipole refinement of com­pound 2 with 0.05 e Å−3 contours. Red contours are positive, blue are negative and dashed grey are zero.
PMC9899513
c-79-00043-fig2.jpg
0.559737
10a53f58db6a4941aecda1b9d2f1cc6a
The mol­ecular structure of com­pound 2, showing 50% probability displacement ellipsoids.
PMC9899513
c-79-00043-fig3.jpg
0.623832
2188dea46e004177b18c79a970624e63
The chalcogen-bonded chains of com­pound 2 propagating along the ac diagonal.
PMC9899513
c-79-00043-fig4.jpg
0.528869
524604ba77bd4365a2702d3bc5a7fa8c
The N⋯Se and O⋯Se chalcogen-bonding inter­actions and π–π stacking inter­actions in the structure of com­pound 2.
PMC9899513
c-79-00043-fig5.jpg
0.501468
f570cf5e0a6646a396fda375c3f5b589
Partial packing diagram of com­pound 2, showing the three-dimensional network built up of chalcogen-bonding inter­actions and π–π stacking inter­actions, viewed parallel to the (010) plane.
PMC9899513
c-79-00043-fig6.jpg
0.612432
5bdfdf43a1a84bbdbf3c510c94563f09
The mol­ecular structure of 2-Me+ iodide, showing 50% probability displacement ellipsoids.
PMC9899513
c-79-00043-fig7.jpg
0.461878
3bcba1d713974961a041920b30b938c1
The π–π stacking inter­actions in the structure of 2-Me+ iodide.
PMC9899513
c-79-00043-fig8.jpg
0.529147
6340ba922e614812857facfeaed33a31
The mol­ecular structure of 2-Me+ tosyl­ate trihydrate, showing 50% probability displacement ellipsoids.
PMC9899513
c-79-00043-fig9.jpg
0.44623
f3ec8c64a0f34d5e8669940f37485c2b
Growth curves of S. costatum (A) and P. globosa (B) in +P and −P groups. Error bars show the mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between +P group and −P group according to a two-way ANOVA with a post-hoc test (**p < 0.01, *p < 0.05).
PMC9899818
fmicb-14-1085176-g001.jpg
0.436982
1654500703d0439382d341a8a4041bb8
Maximum photochemistry efficiency (Fv/Fm) of S. costatum (A) and P. globosa (B) in +P and −P groups. Error bars show the mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between +P group and −P group according to a two-way ANOVA with a post-hoc test (**p < 0.01, *p < 0.05).
PMC9899818
fmicb-14-1085176-g002.jpg
0.502269
3c69c8a9662d4551953a091162d2aa9f
The maximum electron transport rate ETRmax (A: S. costatum, B: P. globosa), light-harvesting efficiency alpha (C: S. costatum, D: P. globosa), and the point of light saturation Ik (E: S. costatum, F: P. globosa) in +P and −P groups. Error bars show the mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between +P group and −P group according to a two-way ANOVA with a post-hoc test (**p < 0.01, *p < 0.05).
PMC9899818
fmicb-14-1085176-g003.jpg
0.428221
0a31089f20e04d75b88b812e7e1f0a83
Normalized chlorophyll a fluorescence transient of S. costatum (A) and P. globosa (B) in −P group for various periods of time plotted by the mean of triplicate (Error bars omitted). Differential curves of ΔVt (obtained by subtracting the curve on day 0 from the samples for various periods of time) in S. costatum (C) and P. globosa (D) under P-limited condition.
PMC9899818
fmicb-14-1085176-g004.jpg
0.380287
3551a03f0be1452f81ed16994054a565
Photosynthetic parameters (relative to day 0) derived by the JIP-test of S. costatum (A) and P. globosa (B) under P-limited condition. φE0 = ET0/ABS = (Fv/Fm) × (1 – VJ), which is the quantum yield of electron transport to intersystem electron acceptors at t = 0; RE0/RC = (RE0/ET0) – (ET0/RC), denoting the reduction in end acceptors on the PSI electron acceptor side per RC at t = 0; RC/CS0 = φP0 × (ABS/CS0) × (VJ/M0), reflecting the amount of active PSII RCs per CS at t = 0. Error bars are mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between +P group and − P group according to a two-way ANOVA with a post-hoc test (**p < 0.01, *p < 0.05).
PMC9899818
fmicb-14-1085176-g005.jpg
0.478057
de6684a1060a4290ac978d1c83ccabd0
Normalized QA− reoxidation kinetic curves of S. costatum and P. globosa in +P and −P groups for various periods of time. Fluorescence decay in the absence of 3-(3,4-dichlorophenyl)-1,1-dimethylurea (DCMU) of S. costatum (A) and P. globosa (B). Fluorescence decay in the presence of 20 μM DCMU of S. costatum (C) and P. globosa (D). Each value represents the mean of the results from triplicates (Error bars omitted).
PMC9899818
fmicb-14-1085176-g006.jpg
0.463466
0fff4166f0e04b98afc858b5e9b00a33
Proportion of PSIIX centers of S. costatum (A) and P. globosa (B) in +P and −P groups for various periods of time. Error bars show the mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between +P group and −P group according to a two-way ANOVA with a post-hoc test (**p < 0.01).
PMC9899818
fmicb-14-1085176-g007.jpg
0.441931
e20f52cf866e459aa7033eccc0effad7
Immunoblot analysis of PSII proteins and stained with Coomassie blue (CB). (A) Immunoblot analysis was performed using antibodies specific to D1. (B) Immunoblot analysis was performed using an antibody specific to PsbO. (C) Ratios of the D1 protein level of P-limited cells were semi-quantitated by the immunoblot analysis relative to that of cells on day 1, which was set to 100% for easy comparison. (D) Ratios of the PsbO protein level of P-limited cells were semi-quantitated by the immunoblot analysis relative to that of cells on day 1, which was set to 100% for easy comparison. Error bars show mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between day 1 and other days according to a two-way ANOVA with a post-hoc test (**p < 0.01, *p < 0.05).
PMC9899818
fmicb-14-1085176-g008.jpg
0.503792
c3e2c7b087d54a0abf44973c17f99be3
(A) Immunoblot analysis of D1 protein levels in S. costatum and P. globosa cultures treated with or without lincomycin in +P and −P groups. The blot was stained with Coomassie blue (CB). (B) Ratios of the D1 protein level of S. costatum cells semi-quantitated by the immunoblot analysis relative to that of cells on day 0, which was set to 100% for easy comparison. (C) Ratios of the D1 protein level of P. globosa cells semi-quantitated by the immunoblot analysis relative to that of cells on day 0. (D) Newly synthesized D1 protein (relative to day 0) in S. costatum in +P and −P groups. Newly synthesized D1 in PSII repair was measured as the change in the active D1 content in the absence of lincomycin (PSII repair active) minus the change in the active D1 content in the presence of lincomycin (PSII repair blocked). (E) Newly synthesized D1 protein (relative to day 0) in P. globosa in +P and −P groups. Newly synthesized D1 in PSII repair was measured as the change in the active D1 content in the absence of lincomycin (PSII repair active) minus the change in the active D1 content in the presence of lincomycin (PSII repair blocked). Error bars are mean ± SE of triplicate treatments. Superscripts indicate significant differences according to a two-way ANOVA with a post-hoc test (B,C: day 0 vs. other days; D,E: +P group vs.−P group. **p < 0.01, *p < 0.05).
PMC9899818
fmicb-14-1085176-g009.jpg
0.461478
bd0fef9799d34ae9875ff3418423570e
Non-photochemical quenching (NPQ) capacity of S. costatum (A) and P. globosa (B) in +P and −P groups. Error bars show the mean ± standard error (SE) of triplicate treatments. Superscripts indicate significant differences between +P group and −P group according to a two-way ANOVA with a post-hoc test (*p < 0.05).
PMC9899818
fmicb-14-1085176-g010.jpg
0.452224
dbcd74a96c5340e3ad1386a3db940bf8
The photosynthetic responses of S. costatum (red line) and P. globosa (blue line) under P limitation (the thickness of the line indicates the strength of the effect).
PMC9899818
fmicb-14-1085176-g011.jpg
0.491391
a0fede4f98fd4922b999e1616ee3a23a
Perioperative course of (A) bio-ADM and (B) penKid. n, number of patients; S1, prior to surgery; S2, intraoperatively at the end of cardiopulmonary bypass; S3, at ICU-admission; S4, 24 h after surgery; and S5, 48 h after surgery; bio-ADM, bioactive Adrenomedullin; penKid, proenkephalin A.
PMC9900105
fcvm-09-1017867-g0001.jpg
0.487439
11ac3bc1388f4f84a1f4a844c09f53ae
AUC/ROC analysis of intraoperative levels of penKid and EuroSCORE II alone (S2) and a combination of penKid (S2) with preoperative EuroSCORE II for the combined outcome “ICU length of stay >1 day or in-hospital death.” AUC/ROC, area under the curve/receiver operating characteristics; penKid, proenkephalin A; S2, intraoperatively at the end of cardiopulmonary bypass; EuroSCORE II, preoperative European System for Cardiac Operative Risk Evaluation Score II.
PMC9900105
fcvm-09-1017867-g0002.jpg
0.38816
253e1f05024d4c82a161987c6877668d
Association of proenkephalin A (penKid) with acute kidney injury (AKI). (A) Comparison of penKid levels in patients with/without AKI. (B) penKid levels in patients by Acute Kidney Injury Network (AKIN) class. n, number of patients; S1, prior to surgery; S2, intraoperatively at the end of cardiopulmonary bypass; S3, at ICU admission; S4, 24 h after surgery; and S5, 48 h after surgery.
PMC9900105
fcvm-09-1017867-g0003.jpg
0.434503
6cd2cdfeb6d5431ebefde991a0400bf3
Association of bioactive Adrenomedullin (bio-ADM) and proenkephalin A (penKid) with the combined endpoint “in-hospital mortality, patients receiving mechanical ventilation support, re-intubation, post-surgery infection and wound infections” (A) bio-ADM and (B) penKid; n, number of patients; S1, prior to surgery; S2, intraoperatively at the end of cardiopulmonary bypass; S3, at ICU admission; S4, 24 h after surgery; and S5, 48 h after surgery.
PMC9900105
fcvm-09-1017867-g0004.jpg
0.424098
1c18cbb5230c4aafbb74508023e3d27c
MiSL predicts ACC1 as a metabolic dependency for mutant IDH1, but not mutant IDH2. A, Schematic showing MiSL algorithm and prediction of ACC1 (ACACA) as a potential synthetic lethal partner of mutant IDH1 across pan-cancer. CNA, copy-number alteration. B,ACC1 mRNA expression comparing mutated IDH1 and IDH2 AML vs. wild-type in The Cancer Genome Atlas based on RNA sequencing data. Differences in expression were compared with Student t test with P values as shown. C, Validation of ACC1-specific hairpins on protein expression by Western blot (left) with quantification (right). Experiment was performed 3 independent times. *, P < 0.05. RFU, relative fluorescence unit. D, ACC1-specific hairpins with mRNA quantified by TaqMan qPCR compared with nontargeting shRNA. Experiment was performed in triplicate 2 independent times; ***, P < 0.001; **, P < 0.01; Student t test. E, Knockdown of ACC1 using shRNA#1 (left) or shRNA#2 (right) in THP-1 cells expressing IDH1 R132H (mIDH1) or IDH2 R140Q (mIDH2) under dox-induced promoter in lipid-depleted media. The number of viable RFP+GFP+ double-positive cells at day 10 was enumerated relative to fluorescent counting beads. RFP tracks integrated ACC1-specific shRNA hairpin; eGFP tracks mutant protein after dox induction. This experiment was performed 3 times with a representative graph shown. Bars represent standard deviation. ***, P < 0.001; **, P < 0.01; *, P < 0.05; Student t test performed on 4 sorted biological replicates for each transduction group. F, Similar experiment using wild-type IDH cells. ACC1 was knocked down using shRNA#1 or shRNA#2 in THP-1 cells expressing IDH1 or IDH2 wild-type proteins under dox-induced promoter in lipid-depleted media. As in D, the number of viable RFP+GFP+ double-positive cells at day 10 was enumerated relative to fluorescent counting beads. RFP tracks integrated ACC1-specific shRNA hairpin; eGFP tracks wild-type protein after doxycycline induction. This experiment was performed 3 times with a representative shown. Bars represent standard deviation. NS, nonsignificant; NT, nontargeting shRNA.
PMC9900324
496fig1.jpg
0.46521
7a078156b26a410eb92b8deea21a75ca
IDH1-mutant AML associated with decreased single-chain fatty acid metabolites. A, Principal component analysis of extracted nonpolar metabolites identified by mass spectrometry in negative ion mode from THP-1 cells cultured in low serum media following induction of mIDH1 R132H, mIDH2 R140Q, wild-type IDH1, or wild-type IDH2. B, Heat map showing differentially abundant nonpolar lipid species globally decreased in IDH1 R132H THP-1 cells (+dox) compared with IDH2 R140Q (+dox) vs. no-dox controls. C, Schematic showing metabolomics protocol for extracting nonpolar lipid species using organic solvent and derivatization of polar metabolites with O-benzylhydroxylamine and dansyl-tags from primary cell AML extracts. Putative metabolite identifications based on mass/charge ratio and retention time were determined from the Human Metabolome Database, Lipid Annotator software, and a validated in-house lipidomics fragmentation library. HMDB, Human Metabolome Database. D, Heat map showing clustering of primary AML samples based on differentially abundant metabolites (all metabolites, both polar and nonpolar). mIDH1 samples are shown in red. CB, cord blood. E, Volcano plot highlighting significant differentially abundant metabolites in healthy IDH1 wild-type CD34+ progenitors vs. mIDH1 AML. Red dashed line indicates a nonadjusted P value threshold of 0.05. F, Volcano plot highlighting significant differentially abundant metabolites in 6 × mIDH1 vs. 5 × mIDH2 AML samples. Dashed line indicates a nonadjusted P value threshold of 0.05. G, Representative examples of distinct lipid metabolites with decreased abundance in IDH1-mutant vs. IDH2-mutant AML and wild-type AML; x-axis shows metabolite abundance. Bars represent standard error of independent samples. All P values represent comparison with wild-type. LPE, lysophosphatidylethanolamine; MG, monoacylglycerolipids; NS, nonsignificant; WT, wild-type.
PMC9900324
496fig2.jpg
0.435321
a1b432c158354594baf45e431d95c5a4
IDH1 mutation is linked to defective reductive carboxylation, increased fatty acid consumption, and decreased NADPH. A, Schematic and graph of flux studies showing the percentage of labeled M2 glycerol-3-phosphate derived from 13C[1,2] labeled glucose (2 of 6 carbons as heavy isotope) across THP-1 cells induced to express mIDH1 vs. IDH1 wild-type compared with mIDH2 vs. IDH2 wild-type. Glucose was added to media in normoxia over 13 hours. Schematic indicates M2 isotopolog of glycerol-3-phosphate de novo synthesis directly from glycolysis utilizing labeled glucose rather than the oxidative pentose phosphate pathway (M1 isotopolog). A two-tailed unpaired Student t test was used to compare differences between groups. This experiment was performed with 6 cell pellets for each sample blinded and randomized on each LC-MS run. DHAP, dihydoxyacetone phosphate; G-6-P, glucose-6-phosphate. B, Decreased reductive carboxylation of mIDH1 compared with IDH1 wild-type and mIDH2 expressed in THP-1 cells measured by the percentage of M5 citrate isotopolog obtained from U-13C5 glutamine labeling in 2% hypoxia over 16 hours, as shown in the schematic. The last bar shows mIDH1 cells cultured in the presence of 10 μmol/L ivosidenib added prior to adding a label. This experiment was performed with 6 cell pellets for each sample blinded and randomized on each LC-MS run. ****, P < 0.0001; Student t test. C, Column graph showing FC increase in acylcarnitine metabolites after induction of mIDH1 (+dox) vs. wild-type (−dox) in comparison with mIDH2 (+dox) vs. IDH2 wild-type (−dox) in THP-1 cells as measured by LC-MS. Student t test is used to compare groups. D, Column graph showing mean fatty acid β-oxidation as measured by the etomoxir (ETO)-sensitive component of oxygen consumption in pmole/min/105 cells in mIDH1 vs. IDH1 wild-type, mIDH2 and IDH2 wild-type in THP-1 cells measured on Seahorse analyzer (3 independent experiments). E, Column graph showing the percentage of b-oxidation of total oxygen consumption (etomoxir-sensitive component) in mIDH1 and mIDH2 before and after the addition of the ACC1 inhibitor 100 nmol/L ND-646. A representative experiment is shown. Statistics represent paired t test with n = 4 replicates. F, Column graph showing percentage of etomoxir-sensitive component in mIDH1 before and after the addition of 10 μmol/L ivosidenib. P = nonsignificant, Student t test, 4 replicates. G, Western blot showing phospho-AMPK (pAMPK) on threonine 172 in THP-1 cells induced with mIDH1, mIDH2, and wild-type counterparts in lipid-replete conditions. Comparison with total AMPK α isoform and Actin is shown in panels below. H, NADPH levels measured in identical 2 × 106 viable cell pellets of mIDH1, mIDH2, or wild-type primary AML blasts vs. normal CD34+ cells grown in culture for 48 hours in normoxia. Symbols indicate individual sample values. Student t test is used to compare groups. **, P < 0.01.I, Schematic summarizing mIDH1-induced mechanisms impacting lipid metabolism. Pathways involved directly in phospholipid synthesis that are perturbed by mIDH1 are shown in red. Thin arrows indicate reduced flux, and thick arrows indicate preserved or increased flux. Black arrows indicate pathways not measurably affected by mIDH1. Mechanistic causes for aberrant lipid metabolism identified in our study include (i) reduced carbon flux arising from defective reductive carboxylation by mutant IDH1 hetero/homodimers exacerbated in relative marrow hypoxia and mitochondrial stress; (ii) cumulative NADP(H) decrease by neomorphic reverse αKG to 2HG reaction; (iii) ongoing depletion of NADPH by residual reductive carboxylation of glutamine; (iv) insufficient NADPH replenishment by impaired forward reaction akin to TCA cycle; (v) increased fatty acid β-oxidation with a concomitant increase in acylcarnitines that is sensitive to ACC1 inhibition, but not ivosidenib; and (vi) decreased AMPK phosphorylation, indicating an AMPK-independent mechanism for enhanced oxidative phosphorylation of fatty acids. In contrast, the flux of glucose to produce glycerol-3-phosphate, the building block for the glycerol component of glycerolipids, is not impaired but rather is upregulated in mIDH1 compared with mIDH2, shown in black. This would suggest de novo synthesis of glycerol from glucose is not the major cause of defective lipid species in mIDH1 AML. Similarly, static levels of citric acid cycle metabolites are decreased to similar levels in both mIDH1 and mIDH2 AML, as shown in Supplementary figures. Blockade of 2HG production by ivosidenib is not sufficient for restoring defective reductive carboxylation or abrogating β-oxidation. NS, nonsignificant; WT, wild-type.
PMC9900324
496fig3.jpg
0.427904
9038657254624cd79d6a78ba4081c44b
Lipid-free diet causes decreased growth of mIDH1 AML. A, Growth of mIDH1 vs. mIDH2 THP-1 cells in lipid-stripped serum over 10 days shown as a relative FC compared with IDH1 wild-type cells. Growth was measured using Presto-Blue cell viability. Bars represent standard deviation of 4 replicates in a representative experiment performed 3 times. B, Schematic showing change in diet to sucrose-rich lipid-free diet at 10 weeks after engraftment of human AML. C, Triglyceride levels measured in NSG mice after 6 weeks of lipid-free dietary supplementation. D–H, Human CD33+ engraftment of mIDH1 AML (D, E), mIDH2 AML (F, G), and normal CD34+ hematopoietic stem/progenitor cells (H) after 6 weeks in mice treated with lipid-free compared with normal rodent diet. Bars represent standard error. Mann–Whitney U test was used to compare engraftments with P values as shown. NS, nonsignificant.
PMC9900324
496fig4.jpg
0.42905
a80418bc4e114737bb7803acc532fa12
ACC1 is a potential target in IDH1-mutant solid tumors. A, Growth of HT-1080 tumors over 21 days after knockdown of ACC1 measured by intravital imaging of shRNA-transduced luciferase+ cells. B,In vivo imaging of tumors from the same experiment. C, Colony assays quantified by crystal violet showing decreased colonies in mIDH1 with ACC1 knockdown compared with wild-type in lipid-replete conditions. D, Column graphs showing mean number of colonies after seeding 2,500 cells per well; bars represent standard error of the mean from 2 independent experiments. E,In vivo growth of IDH1 wild-type HT-1080 cells after CRISPR–Cas9 correction. Plots show median tumor growth after 25 days. P = nonsignificant; Mann–Whitney U test. F, Ki-67 IHC stain of explanted HT-1080 (5 tumors × 10 fields of view) after ACC1 knockdown compared with control with Student t test. Right, quantification of the Ki-67 proliferation index in tumors with ACC1 knockdown vs. control. G, Summary bar graph of 5 tumors × 10 fields of view showing increased apoptosis in ACC1 knockdown explants as measured by IHC staining of cleaved caspase-3 in explanted tumors. H, Kaplan–Meier log-rank survival curve for NSG mice transplanted with HT-1080 cells comparing ACC1 knockdown (pink) to nontargeting control (black) ± ivosidenib (ivo) treatment 25 mg/kg/day (gray, purple) or vehicle given by oral gavage for 50 days. Mice eventually succumbed by 72 days in all treatment groups due to increased tumor growth requiring euthanasia. P values represent log-rank Mantel–Cox test between groups as indicated. I, Bar graph summarizing total flux (photons per second) based on intravital imaging at day 21 for the same experiment. Student t test with indicated P values assessed statistical significance. J, Kaplan–Meier log-rank survival curve for NSG mice transplanted with HT-1080 cells and fed lipid-free vs. normal (lipid-replete) rodent diet. As in the previous experiment, mice eventually succumbed by day 70 in all treatment groups due to increased tumor growth. P values represent log-rank Mantel–Cox test between groups as indicated. kd, knockdown. K, Bar graph summarizing tumor size at 21 days after engraftment as measured by calipers. NS, nonsignificant; NT, nontargeting; WT, wild-type.
PMC9900324
496fig5.jpg
0.440029
ad3405413d24491fb2e13b6c4ba871f5
ACC1 selective inhibitors have activity in mIDH1 cancers. A, Viability of HT-1080 mIDH1 R132C cells after exposure to 4 μmol/L TOFA for 72 hours measured by DAPI-negative cell population. B, Representative images of wild-type IDH1 reversion HT-1080 cells cultured at low density in 4% lipid-depleted serum or IDH1 R132C parental cells or IDH1 R132C treated with 10 μmol/L ivosidenib. Note that cells with IDH1 R132C mutation form ultrathin adherent elongated spindle-like cells. This morphology change was not reversed by coculture with ivosidenib. C, Measurement of 2HG in the supernatant of parental mIHD1 HT-1080 cells but undetectable in wild-type IDH1 reversion HT-1080 cells (clone #65 is shown as representative clone) after 72 hours. D, Total abundance of lysophospholipids (LPC + LPE) measured by LC-MS in mIDH1 vs. WT HT-1080 cultured in lipid-depleted media. AUP, area under the peak. E, Growth curves of mIDH1 vs. wild-type revertant HT-1080 in lipid-replete (left) vs. lipid-depleted (right) serum over 7 days. F, FC decrease in the number of live IDH1 R132C HT-1080 cells compared with IDH1 reversion wild-type HT-1080 cells after 96-hour exposure to the ACC1 inhibitor TOFA compared with DMSO vehicle. Graph shows mean of 3 independent experiments. G, Western blot showing increased phosphorylated ACC1 Serine 79 after treatment of HT-1080 wild-type cells with increasing concentration of AICAR. H, FC decrease in the number of live IDH1 R132C HT-1080 cells after 96-hour exposure to AICAR vs. H2O vehicle in comparison with IDH1 reversion wild-type HT-1080 cells under same conditions. Graph is a summary of 3 independent experiments. I and J, Primary mIDH1 AML cells isolated by flow cytometry from a patient at relapse (I) and a de novo patient (J) are sensitive to 10 μmol/L TOFA over 72 hours, but cytotoxicity is not reversed by 10 μmol/L ivosidenib. ***, P < 0.001;**, P < 0.01; *, P < 0.05. RFU, relative fluorescence unit. K, Summary of TOFA IC50 at 72 hours after in vitro treatment of mIDH1, mIDH2, and IDH wild-type primary AML blasts cultured in low serum media. P value reflects nonparametric two-tailed comparison between groups; bars represent median IC50 in μmol/L. L, Engraftment of mIDH1 AML at baseline and after 30 days of treatment with either vehicle or 50 mg/kg TOFA given by daily intraperitoneal injection. P = 0.007, paired t test, treated vs. baseline. PDx, patient-derived xenograft; WT, wild-type.
PMC9900324
496fig6.jpg
0.464883
4b6df81810a44de69ab3a62e8b916fdb
The ACC1-selective inhibitor ND-646 can overcome venetoclax resistance. A, FC decrease in the number of live IDH1 R132C HT-1080 cells compared with IDH1 reversion wild-type (WT) HT-1080 cells after 96 hours exposure to the ACC1 inhibitor ND-646 compared with DMSO vehicle. Graph shows mean of 3 independent experiments. B, Parental IDH1 R132C HT-1080 cells were treated with increasing concentrations of the ACC1/2 inhibitor ND-646 ± 10 μmol/L ivosidenib. Cell viability was measured after 5 days in 4% lipid-stripped serum culture. This experiment was performed 3 times with a representative experiment shown. Bars represent standard deviation of technical replicates. No statistical differences were observed between ivosidenib-treated cells vs. untreated. RFU, relative fluorescence unit. C, Baseline and posttreatment bone marrow (BM) engraftment levels of human CD45+CD33+ mIDH1 AML exposed to ABT-199 50 mg/kg or ABT-199 50 mg/kg in combination with the selective ACC1 inhibitor ND-646 100 mg/kg for 7 days by oral gavage. P values represent the nonparametric Mann–Whitney U test. D, Peripheral blood (PB) engraftment levels of mIDH1 primary AML after treatment for 14 days with either vehicle, ABT-199 50 mg/kg, or ABT-199 in combination with ND-646. E, Bone marrow engraftment levels of the same experiment in D. P values represent the nonparametric Mann–Whitney U test for D and E. NS, nonsignificant; PDx, patient-derived xenograft.
PMC9900324
496fig7.jpg
0.450355
51ab4626f1214d80a3e2710eebd6def2
Spatio-temporal and social orientation changes along the AD continuum.Mean efficiency scores (ES) of CN (N= 16, blue), MCI (N = 23, green) and AD dementia participants (N = 12, red) for the orientation task in all domains (A), the orientation task in the domains of space and time (spatio-temporal) (B), the orientation task in person (social) (C), and the lexical control task in all domains (D). Significant CN-MCI differences were found in all domain orientation (A; P < 0.05), and space and time orientation (B; P < 0.001). Significant MCI-AD dementia and CN-AD dementia differences were found in all domains, spatio-temporal and social orientation task ES (A, B, C; P < 0.001), as well as in the lexical task (D; P < 0.05). Statistical significance was estimated using ANOVA and Tukey-Kramer post hoc test.
PMC9900945
nihpp-2023.01.25.525587v1-f0001.jpg
0.388254
9fa8394f22a048f8906184615984e58e
Changes in spatio-temporal and social orientation activity in AD.(A) Spatio-temporal and social pattens of brain activity. Results of GLM analysis exhibiting contrast maps of spatio-temporal (A1) and social (A2) orientation tasks over the lexical control task (All participants, DF=50, P<0.05 FDR corrected, cluster size > 20 voxels). (B, C) Divergent changes in spatio-temporal and social orientation activity. Spatio-temporal (B) and social (C) disorientation contrasted task-evoked activity maps across the three clinical groups: CN greater than MCI participants in spatio-temporal (B1) and social (C1) orientation (DF=38, P<0.05 FDR corrected, cluster size thresholding of 20 voxels); CN greater than AD dementia participants in spatio-temporal (B2) and social (C2) orientation (DF=34, P<0.05 FDR corrected, cluster size thresholding of 20 voxels); MCI greater than AD dementia participants in spatio-temporal (B3) and social (C3) orientation (DF=27, P<0.05 FDR corrected, cluster size thresholding of 20 voxels).
PMC9900945
nihpp-2023.01.25.525587v1-f0002.jpg
0.435466
bc005076d5184a8ab794a8bdb6c69760
Orientation-evoked activity overlaps Default Mode sub-networks differently.(A) Spatio-temporal and social activity and DMN A, B, and C overlap. Delineations of DMN sub-networks (Schaefer et. al, 2018) DMN A (dark), DMN B (medium), DMN C (light) superimposed on maps of spatio-temporal (A1) and social (A2) orientation tasks (Orientation > Lexical control; All participants, DF=50, P<0.05 FDR corrected, cluster size > 20 voxels). (B) The precent of overlap between supra-threshold task-evoked spatio-temporal (B1) and social (B2) maps and DMN subnetworks A, B, and C. Asterisks indicate significant overlap (permutation test, 10,000 iterations). (C) Spatio-temporal and social task-evoked coefficients in DMN A, B, and C ROIs. Mean GLM-derived parameter estimates for social (C2 and 3) and spatio-temporal (C1) orientation (>rest) in significantly overlapping (B) DMN subnetworks (C1 – DMN C - spatio-temporal; C2 –DMN B – social; C3 – DMN C - social) for CN (N= 16, blue), MCI (N = 23, green) and AD dementia (N = 12, red). Significant differences were found between CN and AD dementia participants in DMN A, B, and C, between MCI and AD dementia participants in DMN A and B, and between CN and MCI participants in DMN C (ANOVA and Tukey-Kramer post hoc test, P < 0.05).
PMC9900945
nihpp-2023.01.25.525587v1-f0003.jpg
0.420726
5c85f9f24b93405783586486d84803dc
Mediation models of brain activity, glucose metabolism and orientation performance.(A) Mediation analysis was used to test the hypothesis that changes in FDG-PET uptake (M) across the AD continuum alter the relations between spatio-temporal (B2) and social (B2) orientation-evoked brain activity (X) and orientation task performance (Y). (B) Spatio-temporal and social mediation and DMN A, B, and C overlap. Delineations of DMN sub-networks (Schaefer et. al, 2018) DMN A (dark), DMN B (medium), and DMN C (light) superimposed on maps of spatio-temporal (B1) and social (B2) mediation (P<0.05, FDR-corrected). (C) The precent of overlap between supra-threshold task-evoked spatio-temporal (C1) and social (C2) suprathreshold mediation maps and DMN subnetworks A, B, and C. Asterisks indicate significant overlap (permutation test, 10,000 iterations).
PMC9900945
nihpp-2023.01.25.525587v1-f0004.jpg
0.36937
5e4ef6de9c2e486a86bbe2bc971a7286
Postpartum complications mentioned by women who gave birth in the last 12 months in Arba Minch town, Southern Ethiopia, December 2019.
PMC9901784
pone.0281242.g001.jpg
0.459538
11a695b688284bb98294fb3253d44bac
The plasma level of major elements that have relevance to endothelial function in patients with IS and HVs. (A-D) The levels of pro-inflammatory mediator TNF-α and chemoattractant SDF-1 increased in patients at different phases of ischemic stroke while no differences were observed in the levels of G-CSF and total antioxidant capacity. (E, F) Gradual decreases observed in pro-angiogenic factors, VEGF and PDGF-BB reached significance at day 90 of ischemic stroke. (G-J) Ischemic stroke differentially regulated the levels of angiogenic suppressors in that the levels of endostatin constantly increased after ischemic injury while the levels of thrombospondin-1, and thrombospondin-2 decreased. No change was observed in angiostatin levels. * < 0.05, **P < 0.01, ***P < 0.001 versus HVs (t test or Mann Whitney U test). NS: not significant compared to HVs. G-CSF, granulocyte colony-stimulating factor; HVs, healthy volunteers; IS, ischemic stroke; PDGF-BB, platelet-derived growth factor; SDF-1, stromal cell-derived factor 1; TNF-α, tumor necrosis factor-α; VEGF, vascular endothelial growth factor
PMC9902316
12015_2022_10439_Fig1_HTML.jpg
0.41762
0c39d969383d4896b4f7f0519ad618b8
Schematic diagram of an in vitro model of human BBB and the effect of OEC-CM on BBB integrity and function and actin cytoskeleton organization in HBMECs and OECs. (A) In vitro models of human BBB consisting of astrocytes, pericytes, and HBMECs alone or mixed with OECs. (B, C) TNF-α significantly disrupted BBB integrity and function, as shown by decreases in TEER and concomitant increases in paracellular flux of sodium fluorescein, which were prevented by OEC-CM treatment. (D) Co-treatment with OEC-CM prevented the effects of TNF-α on cytoskeletal reorganization in HBMECs and OECs and decreased stress fiber formation (white arrows). (E) Quantification of stress fiber formation in both cells. Scale bar: 25 μm. *P < 0.05 versus BBB formed by HBMECs or control, #P < 0.05 versus BBB formed by HBMECs exposed to TNF-α, †P < 0.05 versus BBB formed by HBMECs exposed to TNF-α and OEC-CM, φP < 0.05 versus BBB formed by HBMECs and OECs, ψP < 0.05 versus BBB formed by HBMECs and OECs exposed to TNF-α (one-way ANOVA followed by Tukey's post-hoc analysis). BBB, blood–brain barrier; HBMECs, human brain microvascular endothelial cells; OEC-CM, outgrowth endothelial cell-derived conditioned medium; OECs, outgrowth endothelial cells; TNF-α, tumor necrosis factor-α
PMC9902316
12015_2022_10439_Fig2_HTML.jpg
0.389959
a0a0bd06703749c895b5036ea4395c4a
The effect of OEC-CM on HBMEC and OEC functional characteristics and analysis of angiogenesis-related proteins in HBMEC and OEC secretomes and OEC-CM. (A, B) OEC-CM accelerated wound closure in both HBMEC and OECs. (C-E) OEC-CM negated the impact of TNF-α on HBMEC and OEC tubule network. (F, G) Treatments with OEC-CM neutralized the inhibitory effect of TNF-α on HBMEC and OEC adhesion to fibronectin, an extracellular matrix protein. (H, I) Proteome profiling of OEC-CM along with HBMEC and OEC secretomes revealed significant variations in various pro- and anti-angiogenic factors e.g. endothelin-1, MCP-1 and endostatin in OEC-CM. Scale bars = 100 μm. *P < 0.05 versus control, #P < 0.05 versus TNF-α (one-way ANOVA followed by Tukey's post-hoc analysis). HBMECs, human brain microvascular endothelial cells; IL-8, interleukin-8; MCP-1, monocyte chemoattractant protein-1; OEC-CM, outgrowth endothelial cell-derived conditioned medium; OECs, outgrowth endothelial cells; TIMP-1, tissue inhibitors of metalloproteinase-1; TNF-α, tumor necrosis factor-α; uPA, urokinase plasminogen activator
PMC9902316
12015_2022_10439_Fig3_HTML.jpg
0.426387
6d50c706077b48a7b8232d896f1f2a54
Primary wintering regions of the Midcontinent population of greater white-fronted goose (Anser albifrons frontalis) in North America (excluding regions in Mexico). Transmitters were deployed during winters 2016–2018 in the Chenier Plain (Louisiana), Lower Texas Coast, and Rolling/High Plains regions. Geese that made winter movements outside of these defined regions were classified as ‘Other’ regions. Map created using Esri ArcMap (version 10.3.1; www.esri.com).
PMC9902612
41598_2023_28937_Fig1_HTML.jpg
0.454489
66a428ab70b247a985baf8e65a20bd13
Mean effect on habitat-specific transition probability (± 95% credible intervals; log-odds scale) to each of seven landcover types for the specific wintering region selected by 56 greater white-fronted geese during winters 2016–2017 and 2017–2018. Note varying scales of the y-axis on each panel to show increased detail due to small credible intervals. Also, effects were not estimated for transitions that were not observed; for example, no estimate is reported for transitions to corn in Chenier Plains in the upper left panel.
PMC9902612
41598_2023_28937_Fig2_HTML.jpg