[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,080)

Search Parameters:
Keywords = knockout mice

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 3327 KiB  
Article
Increased Virulence of Culicoides Midge Cell-Derived Bluetongue Virus in IFNAR Mice
by Barbara S. Drolet, Lindsey Reister-Hendricks, Christie Mayo, Case Rodgers, David C. Molik and David Scott McVey
Viruses 2024, 16(9), 1474; https://doi.org/10.3390/v16091474 (registering DOI) - 17 Sep 2024
Viewed by 79
Abstract
Bluetongue (BT) is a Culicoides midge-borne hemorrhagic disease affecting cervids and ruminant livestock species, resulting in significant economic losses from animal production and trade restrictions. Experimental animal infections using the α/β interferon receptor knockout IFNAR mouse model and susceptible target species are critical [...] Read more.
Bluetongue (BT) is a Culicoides midge-borne hemorrhagic disease affecting cervids and ruminant livestock species, resulting in significant economic losses from animal production and trade restrictions. Experimental animal infections using the α/β interferon receptor knockout IFNAR mouse model and susceptible target species are critical for understanding viral pathogenesis, virulence, and evaluating vaccines. However, conducting experimental vector-borne transmission studies with the vector itself are logistically difficult and experimentally problematic. Therefore, experimental infections are induced by hypodermic injection with virus typically derived from baby hamster kidney (BHK) cells. Unfortunately, for many U.S. BTV serotypes, it is difficult to replicate the severity of the disease seen in natural, midge-transmitted infections by injecting BHK-derived virus into target host animals. Using the IFNAR BTV murine model, we compared the virulence of traditional BHK cell-derived BTV-17 with C. sonorensis midge (W8) cell-derived BTV-17 to determine whether using cells of the transmission vector would provide an in vitro virulence aspect of vector-transmitted virus. At both low and high doses, mice inoculated with W8-BTV-17 had an earlier onset of viremia, earlier onset and peak of clinical signs, and significantly higher mortality compared to mice inoculated with BHK-BTV-17. Our results suggest using a Culicoides W8 cell-derived inoculum may provide an in vitro vector-enhanced infection to more closely represent disease levels seen in natural midge-transmitted infections while avoiding the logistical and experimental complexity of working with live midges. Full article
Show Figures

Figure 1

Figure 1
<p>Clinical scores of IFNAR mice following inoculation with (<b>A</b>) 10<sup>2</sup> PFU and (<b>B</b>) 10<sup>4</sup> PFU BTV-17 derived from <span class="html-italic">Culicoides</span> W8 cells (red) or baby hamster kidney (BHK) cells (blue). Data points are XY staggered to show individual scores.</p>
Full article ">Figure 2
<p>Mean body weights, as percent of starting weight, in IFNAR mice following inoculation with (<b>A</b>) 10<sup>2</sup> PFU and (<b>B</b>) 10<sup>4</sup> PFU BTV-17 derived from <span class="html-italic">Culicoides</span> W8 cells (red) or baby hamster kidney (BHK) cells (blue). Mean weights of mock-infected negative control mice are shown in gray. Error bars represent the standard error of the mean (SEM; n = 6). Paired <span class="html-italic">t</span>-test was used to determine statistical significance between W8 and BHK treatment groups as indicated (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.005, **** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">Figure 3
<p>Survival curves of IFNAR mice following transdermal inoculation with BTV-17 derived from <span class="html-italic">Culicoides</span> W8 cells (red, solid) or baby hamster kidney (BHK) cells (blue, dashed). (<b>A</b>) Mice inoculated with the 10<sup>2</sup> PFU low dose (Mantel–Haenszel Hazard Ratio = 41.03; Log-rank Mantel–Cox <span class="html-italic">p</span> = 0.0005). (<b>B</b>) Mice inoculated with the 10<sup>4</sup> PFU high dose (Mantel–Haenszel Hazard Ratio = 39.12; Log-rank Mantel–Cox <span class="html-italic">p</span> = 0.0009).</p>
Full article ">Figure 4
<p>IFNAR mouse inoculated with 10<sup>2</sup> PFU W8-BTV-17 euthanized 6 dpi due to a clinical score of 3. Severe edema (over 0.5 mL serosanguinous fluid as shown) in the thoracic cavity and wet abdominal serosal surfaces were observed in all IFNAR mice inoculated with both low (10<sup>2</sup> PFU) and high (10<sup>4</sup> PFU) dose W8-BTV-17 and in three inoculated with the high dose of BHK-BTV-17.</p>
Full article ">Figure 5
<p>Mean virus titers in 50 µL daily blood samples as detected by RT-qPCR in IFNAR mice following challenge with (<b>A</b>) 10<sup>2</sup> PFU or (<b>B</b>) 10<sup>4</sup> PFU BTV-17 derived from either <span class="html-italic">Culicoides</span> W8 cells (red with dots) or baby hamster kidney (BHK) cells (blue with triangles). RT-qPCR cycle threshold (Ct; left Y-axis) and Log<sub>10</sub> virus particle calculations (right Y-axis) based on RNA concentrations [<a href="#B39-viruses-16-01474" class="html-bibr">39</a>]. Error bars represent the standard error of the mean (SEM). Multiple paired <span class="html-italic">t</span>-test was used to determine statistical significance between W8 and BHK treatment groups as indicated (ns, not significant, ** <span class="html-italic">p</span> ≤ 0.01). Missing data for W8-BTV-17 inoculated mice was due to 100% mortality by day 6 (low-dose group) and by day 5 (high-dose group).</p>
Full article ">Figure 6
<p>BTV-17 as detected by RT-qPCR in IFNAR mice necropsy tissue samples following challenge with (<b>A</b>) 10<sup>2</sup> PFU or (<b>B</b>) 10<sup>4</sup> PFU BTV-17 derived from either <span class="html-italic">Culicoides</span> W8 cells (red with dots) or baby hamster kidney (BHK) cells (blue with triangles). Mean RT-qPCR cycle threshold (Ct; left Y-axis) and Log<sub>10</sub> virus particle calculations (right Y-axis) based on RNA concentrations [<a href="#B39-viruses-16-01474" class="html-bibr">39</a>] per mL of tissue homogenate. MLN; mesenteric lymph node. One way ANOVA with Sidak’s multiple comparisons was used to determine statistical significance as indicated (* <span class="html-italic">p</span> ≤ 0.05). Error bars represent the standard error of the mean (SEM, n = 6).</p>
Full article ">Figure 7
<p>Infectious virus titers in tissues of IFNAR mice as detected by plaque assay following transdermal inoculation with (<b>A</b>) 10<sup>2</sup> PFU and (<b>B</b>) 10<sup>4</sup> PFU BTV-17 derived from either <span class="html-italic">Culicoides</span> W8 cells (red with dots) or baby hamster kidney (BHK) cells (blue with triangles). MLN; mesenteric lymph node. One-way ANOVA with Sidak’s multiple comparisons was used to determine statistical significance as indicated (* <span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">
18 pages, 3015 KiB  
Article
The BRD4 Inhibitor I-BET-762 Reduces HO-1 Expression in Macrophages and the Pancreas of Mice
by Ana S. Leal and Karen T. Liby
Int. J. Mol. Sci. 2024, 25(18), 9985; https://doi.org/10.3390/ijms25189985 (registering DOI) - 16 Sep 2024
Viewed by 235
Abstract
In pancreatic cancer, the tumor microenvironment (TME) accounts for up to 90% of the tumor mass. Pancreatitis, characterized by the increased infiltration of macrophages into the pancreas, is a known risk factor for pancreatic cancer. The NRF2 (nuclear factor erythroid 2-related factor 2) [...] Read more.
In pancreatic cancer, the tumor microenvironment (TME) accounts for up to 90% of the tumor mass. Pancreatitis, characterized by the increased infiltration of macrophages into the pancreas, is a known risk factor for pancreatic cancer. The NRF2 (nuclear factor erythroid 2-related factor 2) transcription factor regulates responses to oxidative stress and can promote cancer and chemoresistance. NRF2 also attenuates inflammation through the regulation of macrophage-specific genes. Heme oxygenase 1 (HO-1) is expressed by anti-inflammatory macrophages to degrade heme, and its expression is dependent on NRF2 translocation to the nucleus. In macrophages stimulated with conditioned media from pancreatic cancer cells, HO-1 protein levels increased, which correlated with higher NRF2 expression in the nuclear fraction. Significant differences in macrophage infiltration and HO-1 expression were detected in LSL-KrasG12D/+; Pdx-1-Cre (KC) mice, Nrf2 whole-body knockout (KO) mice and wildtype mice with pancreatitis. Since epigenetic modulation is a mechanism used by tumors to regulate the TME, using small molecules as epigenetic modulators to activate immune recognition is therapeutically desirable. When the bromodomain inhibitor I-BET-762 was used to treat macrophages or mice with pancreatitis, high levels of HO-1 were reduced. This study shows that bromodomain inhibitors can be used to prevent physiological responses to inflammation that promote tumorigenesis. Full article
(This article belongs to the Special Issue Immune Modulation of Macrophages: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>NRF2 (nuclear factor erythroid 2-related factor 2) and HO-1 (heme oxygenase 1) expression correlate with overall survival in patients with pancreatic cancer. (<b>A</b>) <span class="html-italic">HMOX-1</span> and <span class="html-italic">NFE2L2</span> (NRF2) expression was evaluated by RNA sequencing in tumors and adjacent normal tissue of patients with pancreatic cancer. Prognostic value of high vs. low gene expression of <span class="html-italic">HMOX1</span> (<span class="html-italic">n</span> = 73 high vs. 104 low) (<b>B</b>), <span class="html-italic">NFE2L2</span> (<span class="html-italic">n</span> = 123 high vs. 54 low) (<b>C</b>) and the combination (<span class="html-italic">n</span> = 50 high vs. 127 low) (<b>D</b>) in patients with pancreatic cancer (<span class="html-italic">n</span> = 177). Data for all plots were accessed and analyzed using KMPLOT Pan-Cancer RNA-seq data base, selected for pancreatic ductal adenocarcinoma. (<b>E</b>) Immunohistochemical evaluation of HO-1 expression in human pancreatic cancer samples from a tissue microarray; arrows indicate positive cells. Scale bar: 120 μm.</p>
Full article ">Figure 2
<p>Inflammatory stimuli and secreted factors from cancer cells increase expression of HO-1 protein in macrophages. (<b>A</b>) Conditioned media (CM) from PanAsc 2159 cells was obtained by culturing cells at 70% confluence for 24 h. RAW 264.7 macrophage-like cells were treated with IL-4, IL-6, LPS and/or IFNγ at 10 ng/mL or CM for 24 h. (<b>B</b>) Bone marrow-derived macrophages (BMDMs) were stimulated with M-CSF for 5 days, followed by IL-4 or CM for 24 h. The Western blot was cut for simplification; the full blot is shown in <a href="#app1-ijms-25-09985" class="html-app">Supplementary Figure S1A</a>. (<b>C</b>) RAW264.7 cells were treated as described in panel A. Cytoplasmic and nuclear fractions were separated. All samples in panels (<b>A</b>–<b>C</b>) were analyzed by Western blotting. (<b>D</b>) mRNA levels of NRF2 and HO-1 in BMDMs cultured, as described in panel (<b>B</b>).</p>
Full article ">Figure 3
<p>Increased expression of HO-1 in the pancreas of LSL-Kras<sup>G12D/+</sup>; Pdx-1-Cre (KC) mice. (<b>A</b>) Experimental design. Nine-week-old LSL-Kras<sup>G12D/+</sup>; Pdx-1-Cre (KC), wildtype (WT) or NRF2<sup>−/−</sup> (knockout) mice were stimulated with caerulein (75 μg/Kg) to induce pancreatitis, and the pancreases were collected 72 h later, <span class="html-italic">n</span> = 5–10 mice/cohort. (<b>B</b>) Weight of pancreas at time of necropsy. (<b>C</b>) Percentages of CD45+ immune cells and (<b>D</b>) CD45+, CD11b+ macrophages in the pancreas were analyzed by flow cytometry. (<b>E</b>) HO-1 protein levels were determined by Western blot of total pancreas homogenates. (<b>F</b>) Immunohistochemistry was used to confirm the levels of HO-1 and the spatial localization of macrophages (F4/80) in the pancreas. Scale bar: 120 μm. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 4
<p>I-BET-762 reduced HO-1 and CD206 in the pancreas of KC mice. KC mice were stimulated with caerulein, as described in <a href="#ijms-25-09985-f003" class="html-fig">Figure 3</a>. I-BET-762 diet was started 72 h before caerulein stimulation. Mice were euthanized 72 hours (<b>A</b>,<b>B</b>) or 9 weeks (<b>C</b>–<b>E</b>) after caerulein stimulation. Pancreas weights, levels of CD45+ immune cells and CD45+, CD11b+ macrophages in the pancreas were analyzed by flow cytometry 72 h (<b>A</b>) or 9 weeks (<b>C</b>) after caerulein stimulation. Levels of HO-1 and/or CD206 were determined by Western blot in total homogenates of pancreases treated for 72 h (<b>B</b>) or 9 weeks (<b>D</b>) with I-BET-762. (<b>E</b>) Immunohistochemistry was used to confirm the levels of HO-1 and CD206 expression and macrophage (F4/80) localization in the pancreases of KC mice 9 weeks after caerulein stimulation. Scale bar: 120 μm. (<b>F</b>) Quantification of acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) lesions in the pancreas of KC mice 9 weeks after caerulein stimulation, <span class="html-italic">n</span> = 4/group. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 5
<p>I-BET-762 reduced the expression of HO-1 in KPC mice. (<b>A</b>) Experimental design. LSL-Kras<sup>G12D/+</sup>; LSL-Trp53<sup>R172H/+</sup>; Pdx-1-Cre (KPC) mice were treated with I-BET-762 for 8 weeks, <span class="html-italic">n</span> = 9 mice/cohort. (<b>B</b>) Weight of pancreas, percentages of CD45<sup>+</sup> immune cells and CD45<sup>+</sup>, CD11b<sup>+</sup> macrophages were analyzed by flow cytometry in the pancreas. (<b>C</b>) Protein expression of HO-1 was determined by Western blot in total homogenates of pancreas, <span class="html-italic">n</span> = 9. (<b>D</b>) Immunohistochemistry was used to confirm expression of HO-1 and spatial localization of macrophages (F4/80) in the pancreas. Scale bar: 120 μm. * <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">
15 pages, 4324 KiB  
Article
Plasminogen Activation Inhibitor-1 Promotes Resilience to Acute Oxidative Stress in Cerebral Arteries from Females
by Safa and Charles E. Norton
Pharmaceuticals 2024, 17(9), 1210; https://doi.org/10.3390/ph17091210 - 14 Sep 2024
Viewed by 243
Abstract
Plasminogen activation inhibitor-1 (PAI-1) plays a central role in thrombus formation leading to stroke; however, the contributions of PAI-1 to cellular damage in response to reactive oxygen species which are elevated during reperfusion are unknown. Given that PAI-1 can limit apoptosis, we hypothesized [...] Read more.
Plasminogen activation inhibitor-1 (PAI-1) plays a central role in thrombus formation leading to stroke; however, the contributions of PAI-1 to cellular damage in response to reactive oxygen species which are elevated during reperfusion are unknown. Given that PAI-1 can limit apoptosis, we hypothesized that PAI increases the resilience of cerebral arteries to H2O2 (200 µM). Cell death, mitochondrial membrane potential, and mitochondrial ROS production were evaluated in pressurized mouse posterior cerebral arteries from males and females. The effects of pharmacological and genetic inhibition of PAI-1 signaling were evaluated with the inhibitor PAI-039 (10 µM) and PAI-1 knockout mice, respectively. During exposure to H2O2, PCAs from male mice lacking PAI-1 had reduced mitochondrial depolarization and smooth muscle cell death, and PAI-039 increased EC death. In contrast, mitochondrial depolarization and cell death were augmented in female PCAs. With no effect of PAI-1 inhibition on resting mitochondrial ROS production, vessels from female PAI-1 knockout mice had increased mitochondrial ROS generation during H2O2 exposure. During acute exposure to oxidative stress, protein ablation of PAI-1 enhances cell death in posterior cerebral arteries from females while limiting cell death in males. These findings provide important considerations for blood flow restoration during stroke treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of pharmacological inhibition and genetic ablation of PAI-1 on H<sub>2</sub>O<sub>2</sub>-induced cell death in PCAs from males and females. Representative images of Hoechst 33342 dye staining all the nuclei (left), propidium iodide (PI) staining the nuclei of dead cells (middle), and merged image (right) following 50 min exposure to H<sub>2</sub>O<sub>2</sub> in isolated pressurized PCAs from male (<b>A</b>) and female (<b>B</b>) mice. In the upper right panel, the white arrow denotes a SMC nuclei, the yellow arrow denotes an EC nuclei, and the green arrow denotes an adventitial cell nuclei (not counted).</p>
Full article ">Figure 2
<p>H<sub>2</sub>O<sub>2</sub>-induced cell death is greater in SMCs of females in the absence of PAI-1 protein expression. Male and female SMC (<b>A</b>,<b>B</b>) and EC (<b>C</b>,<b>D</b>) death following exposure to H<sub>2</sub>O<sub>2</sub> for 50 min in the control/wildtype PCAs ± the inhibitor of PAI-1 (PAI-039 10 µM) or PAI-KO mice. Individual values with means ± SEM for n = 4–6/group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, female vs. male, * <span class="html-italic">p</span> &lt; 0.05, PAI-039 vs. control, <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. control. <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. PAI-039.</p>
Full article ">Figure 3
<p>PAI-1 expression is greater in brain, but not vessels of males. (<b>A</b>) Representative images of PAI-1 (red) and SMC actin (green) from male and female wildtype and PAI-KO mice. Quantification of mean PAI-1 fluorescence in (<b>B</b>) whole brain slices and (<b>C</b>) regions containing blood vessels identified by SMC actin. Individual values with means ± SEM for n = 4–6/group. <sup>#</sup> <span class="html-italic">p</span> &lt;0.05, female vs. male.</p>
Full article ">Figure 4
<p>PAI-1 attenuates the sustained [Ca<sup>2+</sup>]<sub>i</sub> response induced by H<sub>2</sub>O<sub>2</sub> in females. [Ca<sup>2+</sup>]<sub>i</sub> responses (changes in Fura-2 fluorescence) in PCAs obtained from males (<b>A</b>) and females (<b>B</b>) from wildtype/control mice in the absence and presence of PAI-039, and PAI-KO mice during 50 min exposure to H<sub>2</sub>O<sub>2</sub>, followed by a 30 min wash period. Note in females (<b>B</b>), PAI-039 data overlays control data. (<b>C</b>) The peak changes of [Ca<sup>2+</sup>]<sub>i</sub> for both the male and female mice during the exposure to H<sub>2</sub>O<sub>2.</sub> Individual values and means ± SEM for n = 4–6/group. <sup>#</sup> <span class="html-italic">p</span> &lt;0.05, female vs. male, <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. control. <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. PAI-039.</p>
Full article ">Figure 5
<p>PAI-1 does not alter resting ΔΨ<sub>m</sub>. (<b>A</b>) Red (left) and green (center) JC-1 (5 µM) fluorescence as an index of the resting ΔΨ<sub>m</sub> in an intact pressurized PCA from a male wildtype mouse by evaluating the red/green fluorescence ratio (right). The switch from green to red fluorescence results when the dye is dimerized at a high concentration within the mitochondria. The resting ΔΨ<sub>m</sub> in the (<b>B</b>) male and (<b>C</b>) female PCAs from the control/wildtype and PAI-KO mice. Individual values with means ± SEM for n = 6/group. No significant differences were detected.</p>
Full article ">Figure 6
<p>Genetic deletion of PAI-1 enhances depolarization of ΔΨ<sub>m</sub> to a greater extent in females than males. (<b>A</b>) Changes in ΔΨ<sub>m</sub> (TMRM fluorescence; 10 nM; F/F<sub>0</sub>) during H<sub>2</sub>O<sub>2</sub> (200 µM) exposure in PCAs from control male and PAI-KO male mice. (<b>B</b>) Changes in ΔΨ<sub>m</sub> during H<sub>2</sub>O<sub>2</sub> exposure in PCAs from control female and PAI-KO female mice. (<b>C</b>) Peak changes in ΔΨ<sub>m</sub> during H<sub>2</sub>O<sub>2</sub> exposure in PCAs from males and females (control, PAI-039, PAI-KO). Individual values and means ± SEM for n = 5–7/group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, female vs. male, <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. control. <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. PAI-039.</p>
Full article ">Figure 7
<p>PAI-1 Inhibition does not alter mitochondrial ROS production under baseline conditions. MitoSOX fluorescence accumulation under baseline conditions in male (<b>A</b>) and female (<b>B</b>) control/wildtype, PAI-039-treated, and PAI-KO PCAs. (<b>C</b>) MitoSOX fluorescence rate for PCAs from males and females for each treatment. Individual values and means ± SEM for n = 5–6/group. No significant differences were detected.</p>
Full article ">Figure 8
<p>Genetic deletion of PAI-1 enhances mitochondrial ROS production in females during acute oxidative stress. MitoSOX fluorescence accumulation under baseline conditions in male (<b>A</b>) and female (<b>B</b>) control/wildtype, PAI-039-treated, and PAI-KO PCAs. (<b>C</b>) MitoSOX fluorescence rate for PCAs from males and females for each treatment. Individual values and means ± SEM for n = 4–6/group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, female vs. male, <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, PAI-KO vs. Control.</p>
Full article ">
15 pages, 2629 KiB  
Article
Wall Shear Stress (WSS) Analysis in Atherosclerosis in Partial Ligated Apolipoprotein E Knockout Mouse Model through Computational Fluid Dynamics (CFD)
by Minju Cho, Joon Seup Hwang, Kyeong Ryeol Kim and Jun Ki Kim
Int. J. Mol. Sci. 2024, 25(18), 9877; https://doi.org/10.3390/ijms25189877 (registering DOI) - 12 Sep 2024
Viewed by 301
Abstract
Atherosclerosis involves an inflammatory response due to plaque formation within the arteries, which can lead to ischemic stroke and heart disease. It is one of the leading causes of death worldwide, with various contributing factors such as hyperlipidemia, hypertension, obesity, diabetes, and smoking. [...] Read more.
Atherosclerosis involves an inflammatory response due to plaque formation within the arteries, which can lead to ischemic stroke and heart disease. It is one of the leading causes of death worldwide, with various contributing factors such as hyperlipidemia, hypertension, obesity, diabetes, and smoking. Wall shear stress (WSS) is also known as a contributing factor of the formation of atherosclerotic plaques. Since the causes of atherosclerosis cannot be attributed to a single factor, clearly understanding the mechanisms and causes of its occurrence is crucial for preventing the disease and developing effective treatment strategies. To better understand atherosclerosis and define the correlation between various contributing factors, computational fluid dynamics (CFD) analysis is primarily used. CFD simulates WSS, the frictional force caused by blood flow on the vessel wall with various hemodynamic changes. Using apolipoprotein E knockout (ApoE-KO) mice subjected to partial ligation and a high-fat diet at 1-week, 2-week, and 4-week intervals as an atherosclerosis model, CFD analysis was conducted along with the reconstruction of carotid artery blood flow via magnetic resonance imaging (MRI) and compared to the inflammatory factors and pathological staining. In this experiment, a comparative analysis of the effects of high WSS and low WSS was conducted by comparing the standard deviation of time-averaged wall shear stress (TAWSS) at each point within the vessel wall. As a novel approach, the standard deviation of TAWSS within the vessel was analyzed with the staining results and pathological features. Since the onset of atherosclerosis cannot be explained by a single factor, the aim was to find the correlation between the thickness of atherosclerotic plaques and inflammatory factors through standard deviation analysis. As a result, the gap between low WSS and high WSS widened as the interval between weeks in the atherosclerosis mouse model increased. This finding not only linked the occurrence of atherosclerosis to WSS differences but also provided a connection to the causes of vulnerable plaques. Full article
Show Figures

Figure 1

Figure 1
<p>Scheme of WSS evaluation acquisition of ApoE-KO mice atherosclerosis model. (<b>A</b>) Partially ligated ApoE-KO mice fed a high-fat diet were developed as an atherosclerosis model. To confirm disrupted blood flow, (<b>B</b>) MRI images were acquired, along with (<b>C</b>) H&amp;E, Movat staining, and IF staining images. Based on these results, (<b>D</b>) a 3D reconstruction of the carotid artery was performed to evaluate WSS.</p>
Full article ">Figure 2
<p>Blood flow disrupt confirmation through MRI TOF mode and histological staining of carotid artery. (<b>A</b>) Cross-sectioned MRI image and lateral MRI image of 1- and 4-week partial ligated ApoE-KO mice. Partial ligated LCA are indicated as white arrows in MRI images. (<b>B</b>) The excised sample of 4-week mice and H&amp;E and MOVAT staining images of the cross-sectioned tissue at the partial ligation site (bifurcation), the midpoint, and the RCA without partial ligation as control. (<b>C</b>) H&amp;E staining results of 1-, 2-, and 4-week LCA. Pathological features are marked in the image.</p>
Full article ">Figure 3
<p>IF performed on the bifurcation part of the LCA. The samples were double-stained with VCAM-1 and NF-κB as one set, and CD31 and α-SMA as another set. (<b>A</b>–<b>C</b>) represent the 1-, 2-, and 4-week IF staining results for VCAM-1 (green) and NF-κB (red) in the LCA bifurcation. (<b>D</b>–<b>F</b>) show the staining results for CD31 (red) and α-SMA (green) in the LCA bifurcation, respectively. The arrows and dotted lines in the magnified images of each figure indicate the fluorescence biomarker. The arrows in Figures (<b>A</b>–<b>C</b>) represent NF-κB (red). The arrows in the magnified images of Figures (<b>D</b>–<b>F</b>) represent CD31 (red). For VCAM-1 (green), no significant observations were made, and α-SMA (green) was well visualized in Figures (<b>D</b>–<b>F</b>).</p>
Full article ">Figure 4
<p>A 3D construction of the left carotid artery from the midpoint of LCA to bifurcation of the partial ligation part with WSS evaluation. (<b>A</b>) 3D image of LCA with WSS distribution; the red part refers to high WSS, where the blue part refers to low WSS. (<b>B</b>–<b>E</b>) are maximum WSS graphs for 1-, 2-, and 4-week mice in all groups, respectively.</p>
Full article ">Figure 5
<p>Summary of (<b>A</b>) standard deviation of TAWSS, (<b>B</b>) area, (<b>C</b>) maximum WSS, and (<b>D</b>) fluorescence intensity for LCA and RCA (<span class="html-italic">p</span> value &lt; 0.01 ** and &lt;0.05 *).</p>
Full article ">
25 pages, 25762 KiB  
Article
Intermedin Alleviates Diabetic Cardiomyopathy by Up-Regulating CPT-1β through Activation of the Phosphatidyl Inositol 3 Kinase/Protein Kinase B Signaling Pathway
by Jie Zhao, Ling Han, Ya-Rong Zhang, Shi-Meng Liu, Deng-Ren Ji, Rui Wang, Yan-Rong Yu, Mo-Zhi Jia, San-Bao Chai, Hui-Fang Tang, Wei Huang and Yong-Fen Qi
Pharmaceuticals 2024, 17(9), 1204; https://doi.org/10.3390/ph17091204 - 12 Sep 2024
Viewed by 295
Abstract
Diabetic cardiomyopathy (DCM), one of the most serious long-term consequences of diabetes, is closely associated with myocardial fatty acid metabolism. Carnitine palmitoyltransferase-1β (CPT-1β) is the rate-limiting enzyme responsible for β-oxidation of long-chain fatty acids. Intermedin (IMD) is a pivotal bioactive small molecule peptide, [...] Read more.
Diabetic cardiomyopathy (DCM), one of the most serious long-term consequences of diabetes, is closely associated with myocardial fatty acid metabolism. Carnitine palmitoyltransferase-1β (CPT-1β) is the rate-limiting enzyme responsible for β-oxidation of long-chain fatty acids. Intermedin (IMD) is a pivotal bioactive small molecule peptide, participating in the protection of various cardiovascular diseases. However, the role and underlying mechanisms of IMD in DCM are still unclear. In this study, we investigated whether IMD alleviates DCM via regulating CPT-1β. A rat DCM model was established by having rats to drink fructose water for 12 weeks. A mouse DCM model was induced by feeding mice a high-fat diet for 16 weeks. We showed that IMD and its receptor complexes levels were significantly down-regulated in the cardiac tissues of DCM rats and mice. Reduced expression of IMD was also observed in neonatal rat cardiomyocytes treated with palmitic acid (PA, 300 μM) in vitro. Exogenous and endogenous IMD mitigated cardiac hypertrophy, fibrosis, dysfunction, and lipid accumulation in DCM rats and IMD-transgenic DCM mice, whereas knockout of IMD worsened these pathological processes in IMD-knockout DCM mice. In vitro, IMD alleviated PA-induced cardiomyocyte hypertrophy and cardiac fibroblast activation. We found that CPT-1β enzyme activity, mRNA and protein levels, and acetyl-CoA content were increased in T2DM patients, rats and mice. IMD up-regulated the CPT-1β levels and acetyl-CoA content in T2DM rats and mice. Knockdown of CPT-1β blocked the effects of IMD on increasing acetyl-CoA content and on inhibiting cardiomyocyte hypertrophy and cardiac fibroblast activation. IMD receptor antagonist IMD17–47 and the phosphatidyl inositol 3 kinase (PI3K)/protein kinase B (Akt) inhibitor LY294002 reversed the effects of IMD on up-regulating CPT-1β and acetyl-CoA expression and on inhibiting cardiomyocyte hypertrophy and cardiac fibroblast activation. We revealed that IMD alleviates DCM by up-regulating CPT-1β via calcitonin receptor-like receptor/receptor activity-modifying protein (CRLR/RAMP) receptor complexes and PI3K/Akt signaling. IMD may serve as a potent therapeutic target for the treatment of DCM. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Exogenous IMD alleviates DCM in rats. (<b>A</b>) Hematoxylin–eosin staining of representative heart sections and cardiomyocyte cross-sectional area quantification in rats. Scale bar: 50 μm. <span class="html-italic">n</span> = 4. (<b>B</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in the hearts of diabetic rats. <span class="html-italic">n</span> = 3–4. (<b>C</b>) Sirius red staining of myocardial interstitial and perivascular fibrosis area with representative images and quantification in rats. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>D</b>) Quantitative real-time PCR analysis of <span class="html-italic">Col1a1</span> and <span class="html-italic">Col3a1</span> mRNA expression in DCM rat hearts. <span class="html-italic">n</span> = 3–4. (<b>E</b>) Western blot analysis of Col1a1 and Col3a1 protein levels in DCM rat hearts. <span class="html-italic">n</span> = 3. (<b>F</b>) Representative images and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–6. (<b>G</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 3–4. (<b>H</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 1 Cont.
<p>Exogenous IMD alleviates DCM in rats. (<b>A</b>) Hematoxylin–eosin staining of representative heart sections and cardiomyocyte cross-sectional area quantification in rats. Scale bar: 50 μm. <span class="html-italic">n</span> = 4. (<b>B</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in the hearts of diabetic rats. <span class="html-italic">n</span> = 3–4. (<b>C</b>) Sirius red staining of myocardial interstitial and perivascular fibrosis area with representative images and quantification in rats. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>D</b>) Quantitative real-time PCR analysis of <span class="html-italic">Col1a1</span> and <span class="html-italic">Col3a1</span> mRNA expression in DCM rat hearts. <span class="html-italic">n</span> = 3–4. (<b>E</b>) Western blot analysis of Col1a1 and Col3a1 protein levels in DCM rat hearts. <span class="html-italic">n</span> = 3. (<b>F</b>) Representative images and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–6. (<b>G</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 3–4. (<b>H</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>IMD overexpression alleviates DCM in mice. (<b>A</b>) Representative images of hearts, HW/BW ratio, and HW/TL ratio of diabetic WT and IMDtg mice. Scale bar: 10 mm. <span class="html-italic">n</span> = 6–7. (<b>B</b>) Representative echocardiographic images of diabetic WT and IMDtg mice. (<b>C</b>) Representative pulsed Doppler echocardiography pictures, early diastolic mitral flow velocity (<b>E</b>), late diastolic mitral flow velocity (<b>A</b>), and relative quantification of the mitral E/A ratio of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 4–6. (<b>D</b>) Hematoxylin–eosin staining of representative heart sections and cardiomyocyte cross-sectional area quantification in mice. Scale bar: 50 μm. <span class="html-italic">n</span> = 4. (<b>E</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in the hearts of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 3–4. (<b>F</b>) Sirius red staining of myocardial interstitial and perivascular fibrosis area with representative images and quantification from different mice. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>G</b>) Oil Red O staining of myocardial interstitium with representative images and quantification from different mice. Scale bar: 50 μm. <span class="html-italic">n</span> = 5. (<b>H</b>) Quantitative real-time PCR analysis of <span class="html-italic">Col1a1</span> and <span class="html-italic">Col3a1</span> mRNA expression in the hearts of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 4. (<b>I</b>) Western blot analysis of Col1a1 and Col3a1 protein levels in the hearts of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 3. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significant difference.</p>
Full article ">Figure 3
<p>IMD deficiency exacerbates DCM in mice. (<b>A</b>) Representative images of hearts, the HW/BW ratio, and HW/TL ratio of diabetic WT and IMD<sup>−/−</sup> mice. Scale bar: 10 mm. <span class="html-italic">n</span> = 6–8. (<b>B</b>) Representative echocardiographic images of diabetic WT and IMD<sup>−/−</sup> mice. (<b>C</b>) Representative pulsed Doppler echocardiography pictures, early diastolic mitral flow velocity (<b>E</b>), late diastolic mitral flow velocity (<b>A</b>), and relative quantification of the mitral E/A ratio of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 4–5. (<b>D</b>) Hematoxylin–eosin staining of representative heart sections and cardiomyocyte cross-sectional area quantification in mice. Scale bar: 50 μm. <span class="html-italic">n</span> = 4. (<b>E</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3–4. (<b>F</b>) Sirius red staining of myocardial interstitial and perivascular fibrosis area with representative images and quantification in mice. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>G</b>) Oil red O staining of myocardial interstitium with representative images and quantification from different mice. Scale bar: 50 μm. <span class="html-italic">n</span> = 5. (<b>H</b>) Quantitative real-time PCR analysis of <span class="html-italic">Col1a1</span> and <span class="html-italic">Col3a1</span> mRNA expression in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3–4. (<b>I</b>) Western blot analysis of Col1a1 and Col3a1 protein levels in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significant difference.</p>
Full article ">Figure 4
<p>IMD up-regulates CPT-1β. RNA-sequencing analysis of hearts in mice. (<b>A</b>) Volcano plot showing the transcript expression profiles of differentially expressed genes in the hearts of the WT diabetic mice, respectively. <span class="html-italic">n</span> = 3. (<b>B</b>) KEGG pathway enrichment analysis showing the 20 most significantly enriched signaling pathways for differentially expressed genes in the hearts of WT diabetic mice. <span class="html-italic">n</span> = 3. (<b>C</b>) Heat map of the microarray results showing the 36 (ranked by <span class="html-italic">p</span>-values) differentially expressed fatty acid metabolism genes in hearts from the WT diabetic mice. Red, up-regulated; blue, down-regulated; white, no change. <span class="html-italic">n</span> = 3. (<b>D</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in the hearts of WT diabetic mice and diabetic rats. <span class="html-italic">n</span> = 4. (<b>E</b>) Volcano plot showing the transcript expression profiles of differentially expressed genes in the hearts from the diabetic WT and IMD<sup>−/−</sup> mice, respectively. <span class="html-italic">n</span> = 3. (<b>F</b>) KEGG pathway enrichment analysis showing the 20 most significantly enriched signaling pathways for differentially expressed genes in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>G</b>) Heat map of the microarray results showing the 28 (ranked by <span class="html-italic">p</span>-values) differentially expressed fatty acid metabolism genes in hearts from the diabetic WT and IMD<sup>−/−</sup> mice. Red, up-regulated; blue, down-regulated; white, no change. <span class="html-italic">n</span> = 3. (<b>H</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 4. (<b>I</b>) Heat map of the microarray results showing the 23 (ranked by P-values) differentially expressed fatty acid metabolism genes in hearts from the diabetic WT and IMDtg mice. Red, up-regulated; blue, down-regulated; white, no change. <span class="html-italic">n</span> = 3. (<b>J</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in the hearts of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 4. Data are mean ± SD, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>IMD alleviates DCM by up-regulating CPT-1β. (<b>A</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1a</span> and <span class="html-italic">Cpt1b</span> mRNA expression in NRCMs. <span class="html-italic">n</span> =3. (<b>B</b>) Enzyme-linked immunosorbent assay of plasma CPT-1β activity in T2DM patients and healthy controls. <span class="html-italic">n</span> = 38. (<b>C</b>) Enzyme-linked immunosorbent assay of plasma acetyl-CoA concentration in T2DM patients and healthy controls. <span class="html-italic">n</span> = 38–41. (<b>D</b>) Enzyme-linked immunosorbent assay of serum CPT-1β activity in diabetic rats. <span class="html-italic">n</span> = 6. (<b>E</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in the hearts of diabetic rats. <span class="html-italic">n</span> = 4. (<b>F</b>) Western blot analysis of CPT-1β protein levels in diabetic rat hearts. <span class="html-italic">n</span> = 3. (<b>G</b>) Enzyme-linked immunosorbent assay of serum acetyl-CoA concentration in diabetic rats. <span class="html-italic">n</span> = 5–6. (<b>H</b>) Enzyme-linked immunosorbent assay of serum CPT-1β activity of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 6. (<b>I</b>) Western blot analysis of CPT-1β protein levels in the hearts of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 3. (<b>J</b>) Enzyme-linked immunosorbent assay of serum acetyl-CoA concentration in diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 5–6. (<b>K</b>) Enzyme-linked immunosorbent assay of serum CPT-1β activity of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 6. (<b>L</b>) Western blot analysis of CPT-1β protein levels in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>M</b>) Enzyme-linked immunosorbent assay of serum acetyl-CoA concentration in diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 6. (<b>N</b>) Enzyme-linked immunosorbent assay of NRCM supernatant CPT-1β activity. <span class="html-italic">n</span> = 5–6. (<b>O</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 3–4. (<b>P</b>) Western blot analysis of CPT-1β protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>Q</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in supernatant of NRCMs. <span class="html-italic">n</span> = 4. (<b>R</b>) Representative images treated with CPT-1β siRNA and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–4. (<b>S</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs treated with CPT-1β siRNA. <span class="html-italic">n</span> = 4. (<b>T</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>U</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in supernatant of NRCMs. <span class="html-italic">n</span> = 4–6. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significant difference.</p>
Full article ">Figure 5 Cont.
<p>IMD alleviates DCM by up-regulating CPT-1β. (<b>A</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1a</span> and <span class="html-italic">Cpt1b</span> mRNA expression in NRCMs. <span class="html-italic">n</span> =3. (<b>B</b>) Enzyme-linked immunosorbent assay of plasma CPT-1β activity in T2DM patients and healthy controls. <span class="html-italic">n</span> = 38. (<b>C</b>) Enzyme-linked immunosorbent assay of plasma acetyl-CoA concentration in T2DM patients and healthy controls. <span class="html-italic">n</span> = 38–41. (<b>D</b>) Enzyme-linked immunosorbent assay of serum CPT-1β activity in diabetic rats. <span class="html-italic">n</span> = 6. (<b>E</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in the hearts of diabetic rats. <span class="html-italic">n</span> = 4. (<b>F</b>) Western blot analysis of CPT-1β protein levels in diabetic rat hearts. <span class="html-italic">n</span> = 3. (<b>G</b>) Enzyme-linked immunosorbent assay of serum acetyl-CoA concentration in diabetic rats. <span class="html-italic">n</span> = 5–6. (<b>H</b>) Enzyme-linked immunosorbent assay of serum CPT-1β activity of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 6. (<b>I</b>) Western blot analysis of CPT-1β protein levels in the hearts of diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 3. (<b>J</b>) Enzyme-linked immunosorbent assay of serum acetyl-CoA concentration in diabetic WT and IMDtg mice. <span class="html-italic">n</span> = 5–6. (<b>K</b>) Enzyme-linked immunosorbent assay of serum CPT-1β activity of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 6. (<b>L</b>) Western blot analysis of CPT-1β protein levels in the hearts of diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>M</b>) Enzyme-linked immunosorbent assay of serum acetyl-CoA concentration in diabetic WT and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 6. (<b>N</b>) Enzyme-linked immunosorbent assay of NRCM supernatant CPT-1β activity. <span class="html-italic">n</span> = 5–6. (<b>O</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 3–4. (<b>P</b>) Western blot analysis of CPT-1β protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>Q</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in supernatant of NRCMs. <span class="html-italic">n</span> = 4. (<b>R</b>) Representative images treated with CPT-1β siRNA and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–4. (<b>S</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs treated with CPT-1β siRNA. <span class="html-italic">n</span> = 4. (<b>T</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>U</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in supernatant of NRCMs. <span class="html-italic">n</span> = 4–6. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significant difference.</p>
Full article ">Figure 6
<p>IMD up-regulates CPT-1β via its receptor complex. (<b>A</b>) Enzyme-linked immunosorbent assay of NRCM supernatant CPT-1β activity. <span class="html-italic">n</span> = 5–6. (<b>B</b>) Quantitative real-time PCR analysis of <span class="html-italic">Cpt1b</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 3–4. (<b>C</b>) Western blot analysis of CPT-1β protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>D</b>) Representative images treated with IMD<sub>17–47</sub> and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–5. (<b>E</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 3–4. (<b>F</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. (<b>G</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in the supernatants of NRCMs. <span class="html-italic">n</span> = 4. +: added. −: not added. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>IMD up-regulates CPT-1β via the PI3K/Akt signaling pathway. (<b>A</b>) Western blot analysis of p-AMPK (Thr172), AMPK, p-Akt (Ser473), Akt, p-PKA (Thr197), and PKA protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>B</b>) Western blot analysis of p-AMPK (Thr172) and AMPK protein levels in DCM rat hearts. <span class="html-italic">n</span> = 3. (<b>C</b>) Western blot analysis of p-Akt (Ser473) and Akt protein levels in DCM rat hearts. <span class="html-italic">n</span> = 3. (<b>D</b>) Western blot analysis of p-AMPK (Thr172) and AMPK protein levels in the hearts of diabetic WT, IMDtg, and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>E</b>) Western blot analysis of p-Akt (Ser473) and Akt protein levels in the hearts of diabetic WT, IMDtg, and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>F</b>) Enzyme-linked immunosorbent assay of NRCM supernatant CPT-1β activity. <span class="html-italic">n</span> = 5–6. (<b>G</b>,<b>H</b>) Western blot analysis of CPT-1β protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>I</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in the supernatants of NRCMs. <span class="html-italic">n</span> = 4. (<b>J</b>) Representative images and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–4. (<b>K</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 4. (<b>L</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. +: added. −: not added. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significant difference.</p>
Full article ">Figure 7 Cont.
<p>IMD up-regulates CPT-1β via the PI3K/Akt signaling pathway. (<b>A</b>) Western blot analysis of p-AMPK (Thr172), AMPK, p-Akt (Ser473), Akt, p-PKA (Thr197), and PKA protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>B</b>) Western blot analysis of p-AMPK (Thr172) and AMPK protein levels in DCM rat hearts. <span class="html-italic">n</span> = 3. (<b>C</b>) Western blot analysis of p-Akt (Ser473) and Akt protein levels in DCM rat hearts. <span class="html-italic">n</span> = 3. (<b>D</b>) Western blot analysis of p-AMPK (Thr172) and AMPK protein levels in the hearts of diabetic WT, IMDtg, and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>E</b>) Western blot analysis of p-Akt (Ser473) and Akt protein levels in the hearts of diabetic WT, IMDtg, and IMD<sup>−/−</sup> mice. <span class="html-italic">n</span> = 3. (<b>F</b>) Enzyme-linked immunosorbent assay of NRCM supernatant CPT-1β activity. <span class="html-italic">n</span> = 5–6. (<b>G</b>,<b>H</b>) Western blot analysis of CPT-1β protein levels in NRCMs. <span class="html-italic">n</span> = 3. (<b>I</b>) Enzyme-linked immunosorbent assay of acetyl-CoA concentration in the supernatants of NRCMs. <span class="html-italic">n</span> = 4. (<b>J</b>) Representative images and quantification of surface (μm<sup>2</sup>) in NRCMs analyzed by ImageJ 1.53k. Scale bar: 100 μm, 50 μm. <span class="html-italic">n</span> = 3–4. (<b>K</b>) Quantitative real-time PCR analysis of <span class="html-italic">Nppa</span> and <span class="html-italic">Nppb</span> mRNA expression in NRCMs. <span class="html-italic">n</span> = 4. (<b>L</b>) Representative images and quantification of immunofluorescence staining for αSMA (green), Col1a1 (red) and DAPI (blue) in primary cultured rat cardiac fibroblasts. Merged images are shown. Scale bar: 50 μm. <span class="html-italic">n</span> = 3. +: added. −: not added. Data are mean ± SD, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significant difference.</p>
Full article ">
14 pages, 2186 KiB  
Article
Threonine Phosphorylation and the Yin and Yang of STAT1: Phosphorylation-Dependent Spectrum of STAT1 Functionality in Inflammatory Contexts
by Maha M. Elbrashy, Hozaifa Metwally, Shuhei Sakakibara and Tadamitsu Kishimoto
Cells 2024, 13(18), 1531; https://doi.org/10.3390/cells13181531 - 12 Sep 2024
Viewed by 232
Abstract
Threonine phosphorylation promotes inflammatory functions of STAT1 while restricting its interferon (IFN) signaling in innate immune responses. However, it remains unclear whether the restriction of STAT1-mediated IFN signaling conferred by threonine phosphorylation is a ubiquitous mechanism or one that is context-dependent. To address [...] Read more.
Threonine phosphorylation promotes inflammatory functions of STAT1 while restricting its interferon (IFN) signaling in innate immune responses. However, it remains unclear whether the restriction of STAT1-mediated IFN signaling conferred by threonine phosphorylation is a ubiquitous mechanism or one that is context-dependent. To address this, we utilized pristane-induced lupus, a prototype IFN-driven systemic autoimmune disease model characterized by the production of high-titer autoantibodies against nucleic acid-associated antigens. Through genetic and biochemical assays, we demonstrate that Thr748 phosphorylation is dispensable for STAT1 functionality in pristane-induced lupus. Genetically engineered mice expressing the phospho-deficient threonine 748-to-alanine (T748A) mutant STAT1 exhibited similar survival rates, high titers of anti-dsDNA IgG, and nephritis compared to their wild-type littermates. In sharp contrast, STAT1 deficiency protected mice against pristane-induced lupus, as evidenced by increased survival, low titers of anti-dsDNA IgG, and less severe nephritis in the STAT1 knockout mice compared to their T748A littermates. Our study suggests a phosphorylation-dependent modularity that governs the spectrum of STAT1 functionality in inflammatory contexts: IFN phospho-tyrosine-dependent and inflammatory phospho-threonine-dependent, with Thr748 phosphorylation driving selective inflammatory activities, particularly those not driven by the canonical JAK pathway. From a broader perspective, our findings provide deeper insights into how distinct phosphorylation events shape the combinatorial logic of signaling cassettes, thereby regulating context-dependent responses. Full article
Show Figures

Figure 1

Figure 1
<p>T748A mice exhibited similar survival rates and high titers of autoantibodies compared to their Wt littermates following pristane-induced lupus. (<b>A</b>) Schematic diagram of experimental design; (<b>B</b>) survival rate of pristane-injected Wt and T748A littermates (<span class="html-italic">n</span> = 16 mice per genotype); (<b>C</b>,<b>D</b>) serum levels of anti-dsDNA IgG of naïve and pristane-injected Wt and T748A littermates as measured by ELISA (<span class="html-italic">n</span> = 8 mice per genotype per group). <span class="html-italic">p</span> values are shown as measured by log-rank test (Mantel-Cox) (<b>B</b>), one-way ANOVA with post hoc Tukey’s test (<b>C</b>), or unpaired student’s <span class="html-italic">t</span> test with Welch’s correction (<b>D</b>). NT, non-treated.</p>
Full article ">Figure 2
<p>Thr748 phosphorylation is dispensable for Stat1-mediated cytokines expression and IFN signaling following pristane-induced lupus. (<b>A</b>–<b>G</b>) Splenocytes from pristane-injected Wt and T748A littermates. Total RNA was isolated, and the indicated transcripts were quantified by qRT PCR. Data are presented as medians (<span class="html-italic">n</span> = 3 biological replicates). (<b>H</b>) Splenocytes from naïve and pristane-injected Wt and T748A littermates. Whole-cell lysates were harvested and separated by SDS PAGE. The indicated endogenous proteins were detected by Western blotting analysis. Data show three independent biological replicates per genotype per group. (<b>I</b>) Quantification of mean band intensities of (<b>H</b>). <span class="html-italic">p</span> values are shown as measured by unpaired student’s <span class="html-italic">t</span> test with Welch’s correction (<b>A</b>–<b>G</b>) and one-way ANOVA with post hoc Tukey’s test (<b>I</b>). NT, non-treated.</p>
Full article ">Figure 3
<p>T748A mice exhibited similar albuminuria and glomerulonephritis compared to their Wt littermates following pristane-induced lupus. (<b>A</b>) Measurements of the levels of albumin/creatinine in the urine of pristane-injected Wt and T748A littermates as measured by ELISA. (<b>B</b>) Histopathological analysis of glomerulonephritis. Scale bar, 100 μm. (<b>C</b>) Immunofluorescent analysis of autoantibodies deposition in glomeruli. Scale bar, 50 μm. Data are representative of two independent experiments. <span class="html-italic">n</span> = 8 mice per genotype per group for each experiment. <span class="html-italic">p</span> values are shown as measured by unpaired student’s <span class="html-italic">t</span> test with Welch’s correction (<b>A</b>). NT, non-treated.</p>
Full article ">Figure 4
<p>Thr748 phosphorylation is dispensable for STAT1-mediated pathology in pristane-induced lupus. (<b>A</b>) Survival rate of pristane-injected KO and T748A littermates. (<b>B</b>,<b>C</b>) Serum levels of anti-dsDNA IgG of naïve and pristane-injected KO and T748A littermates as measured by ELISA. (<b>D</b>) Measurements of the levels of albumin/creatinine in urine of pristane-injected KO and T748A littermates as measured by ELISA. (<b>E</b>) Histopathological analysis of glomerulonephritis. Data are representative of two independent experiments. <span class="html-italic">n</span> = 4–8 mice per genotype per group for each experiment. Scale bar, 100 μm. <span class="html-italic">p</span> values are shown as measured by log-rank test (Mantel-Cox) (<b>A</b>), one-way ANOVA with post hoc Tukey’s test (<b>B</b>), or unpaired student’s <span class="html-italic">t</span> test with Welch’s correction (<b>C</b>,<b>D</b>). NT, non-treated.</p>
Full article ">Figure 5
<p>Scheme for a phosphorylation-dependent spectrum of STAT1 functionality in inflammatory contexts: IFN phospho-tyrosine-dependent and inflammatory phospho-threonine-dependent.</p>
Full article ">
21 pages, 1224 KiB  
Review
Towards Targeting Endothelial Rap1B to Overcome Vascular Immunosuppression in Cancer
by Behshid Ghadrdoost Nakhchi, Ramoji Kosuru and Magdalena Chrzanowska
Int. J. Mol. Sci. 2024, 25(18), 9853; https://doi.org/10.3390/ijms25189853 - 12 Sep 2024
Viewed by 422
Abstract
The vascular endothelium, a specialized monolayer of endothelial cells (ECs), is crucial for maintaining vascular homeostasis by controlling the passage of substances and cells. In the tumor microenvironment, Vascular Endothelial Growth Factor A (VEGF-A) drives tumor angiogenesis, leading to endothelial anergy and vascular [...] Read more.
The vascular endothelium, a specialized monolayer of endothelial cells (ECs), is crucial for maintaining vascular homeostasis by controlling the passage of substances and cells. In the tumor microenvironment, Vascular Endothelial Growth Factor A (VEGF-A) drives tumor angiogenesis, leading to endothelial anergy and vascular immunosuppression—a state where ECs resist cytotoxic CD8+ T cell infiltration, hindering immune surveillance. Immunotherapies have shown clinical promise. However, their effectiveness is significantly reduced by tumor EC anergy. Anti-angiogenic treatments aim to normalize tumor vessels and improve immune cell infiltration. Despite their potential, these therapies often cause significant systemic toxicities, necessitating new treatments. The small GTPase Rap1B emerges as a critical regulator of Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) signaling in ECs. Our studies using EC-specific Rap1B knockout mice show that the absence of Rap1B impairs tumor growth, alters vessel morphology, and increases CD8+ T cell infiltration and activation. This indicates that Rap1B mediates VEGF-A’s immunosuppressive effects, making it a promising target for overcoming vascular immunosuppression in cancer. Rap1B shares structural and functional similarities with RAS oncogenes. We propose that targeting Rap1B could enhance therapies’ efficacy while minimizing adverse effects by reversing endothelial anergy. We briefly discuss strategies successfully developed for targeting RAS as a model for developing anti-Rap1 therapies. Full article
Show Figures

Figure 1

Figure 1
<p>Targeting EC Rap1B to overcome VEGF-A-induced vascular immuno-suppression—a model. (<b>A</b>) Rap1B-GTP promotes Vascular Endothelial Growth Factor (VEGF) Receptor 2 (VEGFR2) signaling, inhibits proinflammatory signaling, and is a potential anti-cancer target. (<b>B</b>) Deletion of Rap1B in ECs inhibits tumor growth and promotes leukocyte infiltration (Rap1B<sup>iΔEC</sup> mice). (<b>C</b>) Rap1B mediates VEGF-A-induced suppression of proinflammatory nuclear factor κ-light chain enhancer of activated B cells (NF-κB) signaling, including cell adhesion molecule (CAM) expression, limiting T cell adhesion and recruitment. ICAM—intracellular adhesion molecule; IκB—inhibitor of κB; LFA-1—lymphocyte function associated antigen 1 (integrin αLβ2); TNFR1—tumor necrosis factor receptor 1; VCAM—vascular CAM; VLA-4—very late antigen 4 (integrin α4β1). Signal transduction is indicated by arrows.</p>
Full article ">Figure 2
<p>Strategies for targeting WT and mutant RAS. All structures were visualized using PyMOL, with surface models highlighting the binding interfaces; adapted from [<a href="#B87-ijms-25-09853" class="html-bibr">87</a>]. (<b>A</b>) Direct targeting of GDP-bound KRAS-G12C with covalent inhibitor AMG-510 (sotorasib), binding to the switch II pocket, in orange (PDB: 6OIM). (<b>B</b>) Direct targeting of wild-type (WT) and G12C KRAS with RMC-7977 (PDB: 4OBE). RMC-7977 binds to the switch II groove (SIIG) of RAS; (<b>C</b>) GDP-bound KRAS with the SOS1-mediated nucleotide exchange inhibitor DCAI (PDB: 4DST). The surface model highlights the DCAI pocket in yellow. (<b>D</b>,<b>E</b>). Indirect targeting of RAS: surfaces targeted by inhibitors of SOS (a RAS guanine nucleotide exchange factor, GEF, (<b>D</b>)) or effector protein binding (RAS-binding domain, RBD, (<b>E</b>)) (PDB: 6GJ8). (<b>F</b>) Targeting post-translational modification of RAS with tipifarnib (PDB: 4JV6). This structure shows KRAS in complex with farnesyltransferase and the inhibitor tipifarnib, preventing farnesylation of the HVR within CAAX motif. (<b>G</b>) Allosteric inhibition of Ras by the NS1 monobody (PDB: 5E95). The NS1 binding site is highlighted in purple.</p>
Full article ">
19 pages, 6886 KiB  
Article
GSK-3β in Dendritic Cells Exerts Opposite Functions in Regulating Cross-Priming and Memory CD8 T Cell Responses Independent of β-Catenin
by Chunmei Fu, Jie Wang, Tianle Ma, Congcong Yin, Li Zhou, Björn E. Clausen, Qing-Sheng Mi and Aimin Jiang
Vaccines 2024, 12(9), 1037; https://doi.org/10.3390/vaccines12091037 - 10 Sep 2024
Viewed by 557
Abstract
GSK-3β plays a critical role in regulating the Wnt/β-catenin signaling pathway, and manipulating GSK-3β in dendritic cells (DCs) has been shown to improve the antitumor efficacy of DC vaccines. Since the inhibition of GSK-3β leads to the activation of β-catenin, we hypothesize that [...] Read more.
GSK-3β plays a critical role in regulating the Wnt/β-catenin signaling pathway, and manipulating GSK-3β in dendritic cells (DCs) has been shown to improve the antitumor efficacy of DC vaccines. Since the inhibition of GSK-3β leads to the activation of β-catenin, we hypothesize that blocking GSK-3β in DCs negatively regulates DC-mediated CD8 T cell immunity and antitumor immunity. Using CD11c-GSK-3β−/− conditional knockout mice in which GSK-3β is genetically deleted in CD11c-expressing DCs, we surprisingly found that the deletion of GSK-3β in DCs resulted in increased antitumor immunity, which contradicted our initial expectation of reduced antitumor immunity due to the presumed upregulation of β-catenin in DCs. Indeed, we found by both Western blot and flow cytometry that the deletion of GSK-3β in DCs did not lead to augmented expression of β-catenin protein, suggesting that GSK-3β exerts its function independent of β-catenin. Supporting this notion, our single-cell RNA sequencing (scRNA-seq) analysis revealed that GSK-3β-deficient DCs exhibited distinct gene expression patterns with minimally overlapping differentially expressed genes (DEGs) compared to DCs with activated β-catenin. This suggests that the deletion of GSK-3β in DCs is unlikely to lead to upregulation of β-catenin at the transcriptional level. Consistent with enhanced antitumor immunity, we also found that CD11c-GSK-3β−/− mice exhibited significantly augmented cross-priming of antigen-specific CD8 T cells following DC-targeted vaccines. We further found that the deletion of GSK-3β in DCs completely abrogated memory CD8 T cell responses, suggesting that GSK-3β in DCs also plays a negative role in regulating the differentiation and/or maintenance of memory CD8 T cells. scRNA-seq analysis further revealed that although the deletion of GSK-3β in DCs positively regulated transcriptional programs for effector differentiation and function of primed antigen-specific CD8 T cells in CD11c-GSK-3β−/− mice during the priming phase, it resulted in significantly reduced antigen-specific memory CD8 T cells, consistent with diminished memory responses. Taken together, our data demonstrate that GSK-3β in DCs has opposite functions in regulating cross-priming and memory CD8 T cell responses, and GSK-3β exerts its functions independent of its regulation of β-catenin. These novel insights suggest that targeting GSK-3β in cancer immunotherapies must consider its dual role in CD8 T cell responses. Full article
(This article belongs to the Special Issue Vaccines Targeting Dendritic Cells)
Show Figures

Figure 1

Figure 1
<p>Deletion of GSK-3β in DCs led to augmented antitumor immunity in CD11c-GSK-3β<sup>−/−</sup> mice. WT and CD11c-GSK-3β<sup>−/−</sup> mice (<span class="html-italic">n</span> = 7–9) were inoculated with B16F10 melanoma cells, and tumor sizes were monitored. (<b>A</b>,<b>B</b>) CD11c-GSK-3β<sup>−/−</sup> mice exhibited reduced tumor growth compared to WT mice. Tumor sizes from the day of treatment are shown in (<b>A</b>) and tumor weights at the end of the experiment (day 20) are shown in (<b>B</b>). A linear mixed model (Lme4) was fitted to the data in (<b>A</b>), and ANOVA for the fitted linear mixed model was then performed to determine the difference between groups. Student’s <span class="html-italic">t</span>-tests were used for (<b>B</b>). *** <span class="html-italic">p</span> &lt; 0.001. (<b>C</b>) Photo of the tumors at the day 20 after tumor inoculation. Data are representative of two experiments.</p>
Full article ">Figure 2
<p>GSK-3β<sup>−/−</sup> DCs exhibited different expression profiles from β-catenin<sup>active</sup> DCs by scRNA-seq. DCs sorted from spleens of WT (GSK-3β<sup>Flox/Flox</sup>) and CD11c-GSK-3β<sup>−/−</sup> mice, or from WT (β-catenin <sup>Exon3/Exon3</sup>) and CD11c-β-catenin<sup>active</sup> (CD11c-Cre β-catenin<sup>Exon3/Exon3</sup>), were subjected to scRNA-seq as described. (<b>A</b>) Uniform manifold approximation and projection (UMAP) dimensionality reduction mapping analysis of single-cell gene expression of integrated WT (GSK-3β<sup>Flox/Flox</sup>) and GSK-3β<sup>−/−</sup> DCs, and WT (β-catenin<sup>Exon3/Exon3</sup>) and β-catenin<sup>active</sup> DCs. Each dot represents one single cell. A total of 13 clusters were identified and color-coded as indicated. (<b>B</b>) Bubble plots showing the expression of key markers for pDC, cDC1, cDC2, and MoDCs cells among 13 UMAP clusters. The sizes of dots represent the percentages expressed; the color of dot represents the average expression. (<b>C</b>) Bubble plots depicting expression of top DEGs for UMAP clusters shown in (<b>A</b>). (<b>D</b>) Distribution of cells from WT/GSK-3β<sup>Flox/Flox</sup> and GSK-3β<sup>−/−</sup> (left), or WT/β-catenin<sup>Exon3/Exon3</sup> and β-catenin<sup>active</sup> DCs (right) within each of the 13 clusters as depicted in (<b>A</b>). (<b>E</b>) Venn plot showing the overlap of downregulated DEGs (left) and upregulated DEGs (right) in GSK-3β<sup>−/−</sup> DCs versus WT/GSK-3β<sup>Flox/Flox</sup> DCs (GSK-3β<sup>−/−</sup> vs. WT), and β-catenin<sup>active</sup> and WT/β-catenin<sup>Exon3/Exon3</sup> DCs (β-catenin<sup>active</sup> vs. WT). (<b>F</b>) Volcano plot visualizing expression of DEGs in GSK-3β<sup>−/−</sup> and WT/GSK-3β<sup>Flox/Flox</sup> DCs, and their expression pattern in β-catenin<sup>active</sup> and WT/β-catenin<sup>Exon3/Exon3</sup> DCs. DEGs in GSK-3β<sup>−/−</sup> DCs versus WT/GSK-3β<sup>Flox/Flox</sup> DCs are shown in volcano plot (upper), and expression of downregulated DEGs (lower left) and upregulated DEGs (lower right) in β-catenin<sup>active</sup> and WT/β-catenin<sup>Exon3/Exon3</sup> DCs are analyzed and shown in volcano plots. (<b>G</b>) GO enrichment analysis identifies top regulated biological process pathways in in GSK-3β<sup>−/−</sup> DCs vs. WT/GSK-3β<sup>Flox/Flox</sup> DCs (upper), and β-catenin<sup>active</sup> vs. WT/β-catenin<sup>Exon3/Exon3</sup> DCs (lower).</p>
Full article ">Figure 3
<p>Deletion of GSK-3β in DCs does not upregulate β-catenin. (<b>A</b>,<b>B</b>) GSK-3β<sup>−/−</sup> cDCs express similar levels of β-catenin to WT cDCs. WT and GSK-3β<sup>−/−</sup> splenic cDCs were isolated and subjected to Western blot. (<b>A</b>) Expression of GSK-3α/β, β-catenin, and β-actin by Western blotting is shown. One of three experiments is shown. (<b>B</b>) Statistical analysis of β-catenin expression is shown. The relative expression of β-catenin Western blot intensity relative to that of b-actin loading control was calculated, and the ratios for WT cDCs for each experiment were set at 1.0. (<b>C</b>,<b>D</b>) Deletion of GSK-3β in DCs does not upregulate β-catenin. Histogram overlay of β-catenin expression (<b>C</b>) and mean fluorescence intensity (MFI) of β-catenin expression (<b>D</b>) on gated CD11c<sup>+</sup>Bst2<sup>−</sup> cDCs are shown. Student’s <span class="html-italic">t</span>-test, and NS &gt; 0.05. Data shown are representative of at least three experiments.</p>
Full article ">Figure 4
<p>Deletion of GSK-3β in DCs abrogated memory CD8 T cell responses despite augmented cross-priming. (<b>A</b>,<b>B</b>) Deletion of GSK-3β in DCs led to significantly augmented cross-priming. WT and DC-GSK-3β<sup>−/−</sup> mice (<span class="html-italic">n</span> = 4) were immunized with anti-DEC-205-OVA with CpG following adoptive transfer of naïve CFSE-labeled Thy1.1<sup>+</sup> OTI cells, and cross-priming was examined at day 4 after immunization. (<b>A</b>) The percentages of Thy1.1<sup>+</sup> OTI cells out of total CD8 T cells, and (<b>B</b>) the percentages of IFN-γ<sup>+</sup> cells out of total Thy1.1<sup>+</sup>CD8<sup>+</sup> OTI cells in both spleen and draining LN are shown. (<b>C</b>) CD8 memory responses were abrogated in CD11c-GSK-3β<sup>−/−</sup> mice upon recall. Immunized WT and CD11c-GSK-3β<sup>−/−</sup> mice (<span class="html-italic">n</span> = 4–5) were recalled at day 21 and examined 5 days later. The percentages of Thy1.1<sup>+</sup> OTI cells out of total CD8 T cells are shown. Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. Data shown are representative of at least two experiments.</p>
Full article ">Figure 5
<p>scRNA-seq of OVA-specific CD8 T cells identifies distinct populations and reveals differences between CD8 T cells primed in WT and CD11c-GSK-3β<sup>−/−</sup> mice. WT and CD11c-GSK-3β<sup>−/−</sup> mice adoptively transferred Thy1.1<sup>+</sup> OTI CD8 T cells were immunized with anti-DEC-205-OVA plus CpG. Spleen cells were harvested at day 4 or day 10 after immunization, and FACS-sorted OTI cells were subjected to scRNA-seq as described. (<b>A</b>,<b>B</b>) UMAP-dimensionality reduction mapping analysis of single-cell gene expression data of OTI cells isolated 4 or 10 days following vaccination with ant-DEC-205-OVA. Each dot represents one single cell. A total of 9 clusters were identified and color-coded as indicated. UMAP visualization of single cells from combined OTI cells (<b>A</b>), or OT1 cells from WT or CD11c-GSK-3β<sup>−/−</sup> mice at day 4 and day 10 (<b>B</b>) are shown. (<b>C</b>) Bubble plots depicting expression of top DEGs for UMAP clusters shown in (<b>A</b>). (<b>D</b>) Distribution of OTI cells from either WT or CD11c-GSK-3β<sup>−/−</sup> mice at day 4 or day 10 within each of the 9 clusters as depicted in (<b>A</b>). (<b>E</b>) Bubble plots showing the key signatures for CD8 T cells effector and memory phenotype. (<b>F</b>) Expression of effector markers among the UMAP clusters. Gradient expression levels are color-coded as indicated. (<b>G</b>) Violin plot depicting the module score of gene sets associated with effector on OTI cells from either WT or CD11c-GSK-3β<sup>−/−</sup> mice at day 4 or day 10. *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 (<b>H</b>) Signaling pathways that are significantly downregulated or upregulated in OTI cells primed in CD11c-GSK-3β<sup>−/−</sup> mice compared to OTI cells from WT mice at day 4 and day 10.</p>
Full article ">Figure 6
<p>Schematic representation of GSK-3β’s dual roles in regulating CD8 T cell responses. Inhibition of GSK-3β is generally believed to upregulate β-catenin, leading to increased IL-10 production, which suppresses cross-priming and reduces memory CD8 T cell responses. However, our studies demonstrate that genetic deletion of GSK-3β in CD11c<sup>+</sup> DCs does not result in β-catenin accumulation (activation). Instead, the deletion of GSK-3β in DCs enhances cross-priming of CD8 T cells, as indicated by an increase in effector cells and a higher effector index, based on scRNA-seq analysis. Despite this enhanced cross-priming, memory CD8 T cells are nearly abrogated in CD11c-GSK-3β<sup>−/−</sup> mice, likely due to a significant loss of both effector and memory CD8 T cell populations. Collectively, these findings reveal novel mechanisms by which GSK-3β exerts opposing effects on CD8 T cell responses.</p>
Full article ">
14 pages, 3263 KiB  
Article
Ccn2 Deletion Reduces Cardiac Dysfunction, Oxidative Markers, and Fibrosis Induced by Doxorubicin Administration in Mice
by Antonio Tejera-Muñoz, Marcelino Cortés, Alianet Rodriguez-Rodriguez, Lucia Tejedor-Santamaria, Vanessa Marchant, Sandra Rayego-Mateos, Maria José Gimeno-Longas, Andrew Leask, Tri Q. Nguyen, María Martín, Jose Tuñón, Isabel Rodríguez, Marta Ruiz-Ortega and Raul R. Rodrigues-Díez
Int. J. Mol. Sci. 2024, 25(17), 9617; https://doi.org/10.3390/ijms25179617 - 5 Sep 2024
Viewed by 383
Abstract
Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting [...] Read more.
Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting that its role in the cardiovascular system is not yet fully elucidated. In this study, we aimed to investigate the effects of Ccn2 gene deletion on the progression of acute cardiac injury induced by doxorubicin (DOX), a widely utilized chemotherapeutic agent. To this end, we employed conditional knockout (KO) mice for the Ccn2 gene (CCN2-KO), which were administered DOX and compared to DOX-treated wild-type (WT) control mice. Our findings demonstrated that the ablation of CCN2 ameliorated DOX-induced cardiac dysfunction, as evidenced by improvements in ejection fraction (EF) and fractional shortening (FS) of the left ventricle. Furthermore, DOX-treated CCN2-KO mice exhibited a significant reduction in the gene expression and activation of oxidative stress markers (Hmox1 and Nfe2l2/NRF2) relative to DOX-treated WT controls. Additionally, the deletion of Ccn2 markedly attenuated DOX-induced cardiac fibrosis. Collectively, these results suggest that CCN2 plays a pivotal role in the pathogenesis of DOX-mediated cardiotoxicity by modulating oxidative stress and fibrotic pathways. These findings provide a novel avenue for future investigations to explore the therapeutic potential of targeting CCN2 in the prevention of DOX-induced cardiac dysfunction. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Effects of DOX administration on body weight and cardiac mass in mice. Effects of DOX administration in heart weight (<b>A</b>) and the ratio of heart weight to tibia length (<b>B</b>) in mice. * <span class="html-italic">p</span> &lt; 0.05 vs. Control. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 2
<p>Impact of DOX administration on cardiac function in mice. Changes in values of ejection fraction (EF) (<b>A</b>) and fractional shortening (<b>B</b>) (expressed as percentages) with and without DOX administration in mice. (<b>C</b>) M-mode echocardiography representative images obtained five days post-DOX administration for the assessment of cardiac function (See <a href="#app1-ijms-25-09617" class="html-app">Supplementary Figure S1</a> for enlarged images). * <span class="html-italic">p</span> &lt; 0.05 vs. Control; # <span class="html-italic">p</span> &lt; 0.05 vs. Control + DOX. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 3
<p>Gene expression related to cardiac damage in the hearts of study mice. The expression levels of <span class="html-italic">Myh7</span> (<b>A</b>) in the different experimental groups show a significant increase in both the Control + DOX and CCN2-KO + DOX groups compared to the groups without DOX treatment. For <span class="html-italic">Myh6</span> (<b>B</b>), DOX administration has varying effects depending on the deletion of CCN2, resulting in a significant decrease in the Control group, while this change is not observed in the CCN2-KO group. * <span class="html-italic">p</span> &lt; 0.05 vs. Control; # <span class="html-italic">p</span> &lt; 0.05 vs. Control + DOX. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 4
<p>Gene expression related to oxidative stress in the hearts of studied mice. The expression levels of <span class="html-italic">Hmox1</span> (<b>A</b>) and <span class="html-italic">Nfe2l2</span> (<b>B</b>) in the different experimental groups show a significant increase in the Control + DOX group compared to the Control group without DOX treatment. Additionally, for both genes, DOX administration in the absence of CCN2 results in a significant reduction in the increase compared to the Control + DOX group. * <span class="html-italic">p</span> &lt; 0.05 vs. Control; # <span class="html-italic">p</span> &lt; 0.05 vs. Control + DOX. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 5
<p>Immunohistochemistry (IHC) detection of NRF2 protein activation in the left ventricular tissue of mouse hearts. (<b>A</b>) Representative IHC images showing phospho-NRF2 staining. (<b>B</b>) Quantification of positive staining for phospho-NRF2 (p-NRF2) levels. Five days of DOX administration resulted in significantly elevated phospho-NRF2 levels in the Control group mice. In contrast, the CCN2-KO + DOX group did not show significant changes in protein activation compared to their corresponding CCN2-KO control group. * <span class="html-italic">p</span> &lt; 0.05 vs. Control; # <span class="html-italic">p</span> &lt; 0.05 vs. Control + DOX. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 6
<p>DOX administration increases apoptosis biomarkers in mouse hearts. (<b>A</b>) <span class="html-italic">Bcl2</span>, (<b>B</b>) <span class="html-italic">Bax</span>, and (<b>C</b>) <span class="html-italic">Mcl1</span> levels were measured, showing a significant increase in both Control and CCN2-KO groups after DOX treatment. * <span class="html-italic">p</span> &lt; 0.05 vs. Control. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 7
<p>Cardiac fibrosis in mice assessed using Masson’s trichrome staining of left ventricular (LV) sections. (<b>A</b>) Representative images of the stained sections. (<b>B</b>) Quantitative analysis revealed a significant increase in fibrosis in both DOX-treated groups compared to the untreated groups. However, the Control + DOX group exhibited a greater extent of fibrosis than the CCN2-KO + DOX group. * <span class="html-italic">p</span> &lt; 0.05 vs. Control; # <span class="html-italic">p</span> &lt; 0.05 vs. Control + DOX. <span class="html-italic">n</span> = 6–10.</p>
Full article ">Figure 8
<p>Schematic representation of the experimental model. The diagram outlines the various treatment groups and experimental procedures used in the study, illustrating the administration of DOX and CCN2 deletion applied to the mice.</p>
Full article ">
20 pages, 3168 KiB  
Article
Left Ventricular Systolic Dysfunction in NBCe1-B/C-Knockout Mice
by Clayton T. Brady, Aniko Marshall, Lisa A. Eagler, Thomas M. Pon, Michael E. Duffey, Brian R. Weil, Jennifer K. Lang and Mark D. Parker
Int. J. Mol. Sci. 2024, 25(17), 9610; https://doi.org/10.3390/ijms25179610 - 5 Sep 2024
Viewed by 246
Abstract
Congenital proximal renal tubular acidosis (pRTA) is a rare systemic disease caused by mutations in the SLC4A4 gene that encodes the electrogenic sodium bicarbonate cotransporter, NBCe1. The major NBCe1 protein variants are designated NBCe1-A, NBCe1-B, and NBCe1-C. NBCe1-A expression is kidney-specific, NBCe1-B is [...] Read more.
Congenital proximal renal tubular acidosis (pRTA) is a rare systemic disease caused by mutations in the SLC4A4 gene that encodes the electrogenic sodium bicarbonate cotransporter, NBCe1. The major NBCe1 protein variants are designated NBCe1-A, NBCe1-B, and NBCe1-C. NBCe1-A expression is kidney-specific, NBCe1-B is broadly expressed and is the only NBCe1 variant expressed in the heart, and NBCe1-C is a splice variant of NBCe1-B that is expressed in the brain. No cardiac manifestations have been reported from patients with pRTA, but studies in adult rats with virally induced reduction in cardiac NBCe1-B expression indicate that NBCe1-B loss leads to cardiac hypertrophy and prolonged QT intervals in rodents. NBCe1-null mice die shortly after weaning, so the consequence of congenital, global NBCe1 loss on the heart is unknown. To circumvent this issue, we characterized the cardiac function of NBCe1-B/C-null (KOb/c) mice that survive up to 2 months of age and which, due to the uninterrupted expression of NBCe1-A, do not exhibit the confounding acidemia of the globally null mice. In contrast to the viral knockdown model, cardiac hypertrophy was not present in KOb/c mice as assessed by heart-weight-to-body-weight ratios and cardiomyocyte cross-sectional area. However, echocardiographic analysis revealed reduced left ventricular ejection fraction, and intraventricular pressure–volume measurements demonstrated reduced load-independent contractility. We also observed increased QT length variation in KOb/c mice. Finally, using the calcium indicator Fura-2 AM, we observed a significant reduction in the amplitude of Ca2+ transients in paced KOb/c cardiomyocytes. These data indicate that congenital, global absence of NBCe1-B/C leads to impaired cardiac contractility and increased QT length variation in juvenile mice. It remains to be determined whether the cardiac phenotype in KOb/c mice is influenced by the absence of NBCe1-B/C from neuronal and endocrine tissues. Full article
Show Figures

Figure 1

Figure 1
<p>Structure and expression of NBCe1 major isoforms. (<b>A</b>) An illustration of NBCe1-B protein topology. All NBCe1 isoforms have 14 transmembrane spans (TM1–14), with soluble N-terminal and C-terminal (Nt and Ct) domains located within the cytoplasm. A glycosylated extracellular loop joins TMs 5 and 6. (<b>B</b>) An illustration of sequence differences between NBCe1 isoforms. Due to an alternative upstream promoter that controls NBCe1-B translation, there is a different 85-amino acid (aa) Nt sequence in NBCe1-B (shown in blue) that replaces the first 41 aa residues of NBCe1-A (shown in red). NBCe1-C is identical to NBCe1-B except that the last 46 aa residues of the Ct sequence in NBCe1-B (shown in yellow) are replaced by a different 61 aa sequence (shown in grey) as a consequence of alternative splicing. (<b>C</b>) An illustration of the expression pattern of NBCe1 protein isoforms. The figure was created using BioRender.com.</p>
Full article ">Figure 2
<p>Echocardiography demonstrates impaired left ventricular function in KO<sub>b/c</sub> mice. (<b>A</b>) Representative cross-sectional M-mode images of the left ventricle of WT and KO<sub>b/c</sub> mice between 4–5 weeks of age. (<b>B</b>) Heart rates were titrated to between ~400–500 BPM via isoflurane anesthesia. (<b>C</b>) KO<sub>b/c</sub> mice were found to have significantly greater left ventricle internal diameters during diastole (LVIDd) and systole (LVIDs). (<b>D</b>) KO<sub>b/c</sub> mice also had significantly greater end-diastolic volume (EDV) and end-systolic volume (ESV) than WT mice as calculated from LVID measurements. (<b>E</b>) There was no significant difference in stroke volume between WT and KO<sub>b/c</sub> mice. (<b>F</b>) The fractional shortening of KO<sub>b/c</sub> mice was significantly less than that of WT mice. (<b>G</b>) The ejection fraction of KO<sub>b/c</sub> mice was significantly less than that of WT mice. Data presented as mean ± SEM, n = 11–14 per group. Outliers were defined a priori as any point &gt;2 standard deviations from the mean and were excluded from analysis. WT outliers (<span class="html-italic">n</span>) were excluded from LVIDd (1), LVIDs (1), EDV (1), ESV (1), SV (1), FS (1), and EF (2) data sets. KO<sub>b/c</sub> outliers (<span class="html-italic">n</span>) were excluded from heart rate (1), LVIDd (1), LVIDs (1), EDV (1), ESV (1), SV (2), FS (2), and EF (2) data sets. A significant difference between WT and KO<sub>b/c</sub> groups is indicated in the figure by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 calculated using Student’s unpaired 2-tailed <span class="html-italic">T</span>-test; ns (non-significant).</p>
Full article ">Figure 3
<p>Impaired left ventricular function in KO<sub>b/c</sub> mice is not attributable to differences in left ventricle wall thickness or systemic vascular resistance. During diastole there was no significant difference between the width of the WT and KO<sub>b/c</sub> left ventricle anterior (<b>A</b>) or posterior (<b>B</b>) wall. Similarly, during systole, there was no significant difference between the width of the WT and KO<sub>b/c</sub> left ventricle anterior (<b>C</b>) or posterior (<b>D</b>) wall. (<b>E</b>) There was no significant difference in systolic, diastolic, or mean arterial pressures of awake WT and KO<sub>b/c</sub>. Data presented as mean ± SEM, n = 11–14 per group (panels <b>A</b>–<b>D</b>) or 7–11 per group (panel <b>E</b>). Outliers were defined a priori as any point &gt;2 standard deviations from the mean and were excluded from analysis. WT outliers (<span class="html-italic">n</span>) were excluded from LVAWd (1), LVPWd (1), and LVPWs (1) data sets. KO<sub>b/c</sub> outliers (<span class="html-italic">n</span>) were excluded from LVAWd (1) and LVAWs (1) data sets. Statistical significance calculated using Student’s unpaired 2-tailed <span class="html-italic">T</span>-test; ns (non-significant).</p>
Full article ">Figure 4
<p>Left intraventricular pressure–volume (PV) assessment reveals no significant difference between WT and KO<sub>b/c</sub> mice in load-dependent measures of contractility or relaxation. There was no significant difference between WT and KO<sub>b/c</sub> left ventricular end-systolic pressure (<b>A</b>) or end-diastolic pressure (<b>B</b>). There was no significant difference between WT and KO<sub>b/c</sub> mice in their left ventricular maximum rate of pressure change (dP/dt max, representing load-dependent contractility) (<b>C</b>) or in their minimum rate of pressure change (dP/dt min, representing load-dependent relaxation) (<b>D</b>). Heart rates were titrated to between ~300–500 BPM via isoflurane anesthesia (<b>E</b>). Data presented as mean ± SEM, n = 13–14 per group. Outliers were defined a priori as any point &gt;2 standard deviations from the mean and were excluded from analysis. WT outliers (<span class="html-italic">n</span>) were excluded from end-systolic pressure (1), dP/dt max (1), and heart rate (1) data sets. KO<sub>b/c</sub> outliers (<span class="html-italic">n</span>) were excluded from end-systolic pressure (1), end-diastolic pressure (1), dP/dt max (1), dP/dt min (1), and heart rate (1) data sets. A significant difference between WT and KO<sub>b/c</sub> groups is indicated in the figure by *** <span class="html-italic">p</span> &lt; 0.001 calculated using Student’s unpaired 2-tailed <span class="html-italic">T</span>-test; ns (non-significant).</p>
Full article ">Figure 5
<p>Left intraventricular pressure–volume (PV) assessment during IVC occlusion reveals diminished load-independent contractility in KO<sub>b/c</sub> mice. Representative PV loops obtained in WT (<b>A</b>) and KO<sub>b/c</sub> (<b>B</b>) mice during IVC occlusion used as a preload reduction maneuver to assess load-independent contractility (slope of the end-systolic pressure volume relationship [ESPVR]) and relaxation (slope of the end-diastolic pressure volume relationship [EDPVR]). (<b>C</b>) The slope of the ESPVR was significantly reduced in KO<sub>b/c</sub> mice. (<b>D</b>) The slope of the EDPVR was not significantly different between WT and KO<sub>b/c</sub> mice. (<b>E</b>) Plotting ESPVR against heart rate for individual mice illustrates that ESPVR is independent of heart rate, supporting that although KO<sub>b/c</sub> mice have a slower heart rate than WT during this experiment, this does not account for the observed reduction in their ESPVR. Data presented as mean ± SEM, n = 12–15 per group. Outliers were defined a priori as any point &gt;2 standard deviations from the mean and were excluded from analysis. A single WT outlier was excluded from the EDPVR data set. A single KO<sub>b/c</sub> outlier was excluded from the ESPVR data set. A significant difference between WT and KO<sub>b/c</sub> groups is indicated in the figure by ** <span class="html-italic">p</span> &lt; 0.01 calculated using Student’s unpaired 2-tailed <span class="html-italic">T</span>-test; ns (non-significant).</p>
Full article ">Figure 6
<p>Absence of cardiac hypertrophy in KO<sub>b/c</sub> hearts. (<b>A</b>) Representative low-magnification tiled images, with higher magnified regions of interest (black boxes in low-magnification images), taken of WT and KO<sub>b/c</sub> heart sections stained with H&amp;E. (<b>B</b>) The HW/BW ratio, an index of heart size, was not significantly different between WT and KO<sub>b/c</sub> mice. (<b>C</b>) There was also no significant difference in cross-sectional area between genotypes. Data presented as mean ± SEM, n = 16–18 per group (panel <b>B</b>) or 13–10 per group (panel <b>C</b>). Outliers were defined a priori as any point &gt;2 standard deviations from the mean and were excluded from analysis. A single WT outlier was excluded from the HW/BW ratio data set. A single KO<sub>b/c</sub> outlier was excluded from the HW/BW ratio data set. For panel (<b>B</b>), the statistical significance was calculated using Student’s unpaired 2-tailed <span class="html-italic">T</span>-test. For panel (<b>C</b>), the cross-sectional area of 25–29 cardiomyocytes was measured across 5 images taken around the left ventricle and averaged for each individual mouse, with the statistical significance calculated using hierarchal statistical analysis (nested <span class="html-italic">T</span>-test). ns (non-significant).</p>
Full article ">Figure 7
<p>Increased QT length variation in KO<sub>b/c</sub> mice. Representative average ECG cycles of WT (<b>A</b>) and KO<sub>b/c</sub> (<b>B</b>) mice were created from 5 s segments of Lead-I recordings. The black line represents the average trace, with underlying grey lines representing each individual cycle. This method was applied to 30 s Lead-I recordings of WT and KO<sub>b/c</sub> mice from which QT length and QT length variation were assessed. QT length variation was calculated as the coefficient of variation (SD/mean) across 5 s intervals from a continuous 30 s ECG trace (i.e., 6 × 5 s intervals). (<b>C</b>) Heart rates were titrated to between ~350–500 BPM via isoflurane anesthesia. (<b>D</b>) There was no significant difference between the length of the QT interval in WT and KO<sub>b/c</sub> mice. (<b>E</b>) The QT length variation in KO<sub>b/c</sub> was significantly greater than in WT mice. Data presented as mean ± SEM, n = 11–13 per group. A significant difference between WT and KO<sub>b/c</sub> groups is indicated in the figure by *** <span class="html-italic">p</span> &lt; 0.001 calculated using Student’s unpaired 2-tailed <span class="html-italic">T</span>-test; ns (non-significant).</p>
Full article ">Figure 8
<p>KO<sub>b/c</sub> cardiomyocytes have reduced Ca<sup>2+</sup>-transient amplitude. (<b>A</b>) Representative Ca<sup>2+</sup> transients recorded in individual cardiomyocytes isolated from WT and KO<sub>b/c</sub> mice loaded with the intracellular Ca<sup>2+</sup> indicator Fura-2 AM. Traces represent the average of ~100 consecutive transients recorded in a single cardiomyocyte while paced at 5 Hz. (<b>B</b>) There was no significant difference between WT and KO<sub>b/c</sub> ‘baseline’ F<sub>340/380</sub> ratio. (<b>C</b>) The ‘peak amplitude’ was significantly decreased in KO<sub>b/c</sub> cardiomyocytes. (<b>D</b>) The ‘peak amplitude as % baseline’ (describing the % change from baseline of the Ca<sup>2+</sup> transient) was also significantly decreased in KO<sub>b/c</sub> cardiomyocytes. (<b>E</b>) There was no significant difference between WT and KO<sub>b/c</sub> in ‘time to peak’. (<b>F</b>) There was no significant difference between WT and KO<sub>b/c</sub> in ‘time to 90% baseline’. (<b>G</b>) There was no significant difference between WT and KO<sub>b/c</sub> the Ca<sup>2+</sup> exponential ‘decay constant (tau)’. Data presented as mean ± SEM, n = 8–9 per group with each point representing the mean of 9–12 cells. Outliers were defined a priori as any point &gt;2 standard deviations from the mean and were excluded from analysis. A single WT outlier was excluded from the ‘time to 90% baseline’ data set. A significant difference between WT and KO<sub>b/c</sub> groups is indicated in the figure by * <span class="html-italic">p</span> &lt; 0.05 calculated using hierarchal statistical analysis (nested <span class="html-italic">T</span>-test); ns (non-significant).</p>
Full article ">
22 pages, 10840 KiB  
Article
Murine Retina Outer Plexiform Layer Development and Transcriptome Analysis of Pre-Synapses in Photoreceptors
by Soo-Young Kim, Christine Haewon Park, Bo-Hyun Moon and Gail K. Seabold
Life 2024, 14(9), 1103; https://doi.org/10.3390/life14091103 - 2 Sep 2024
Viewed by 492
Abstract
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited [...] Read more.
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited knowledge about rod versus (vs.) cone synapse development and the associated genes. The transcription factor neural retina leucine zipper (NRL) determines the rod vs. cone photoreceptor cell fate and is critical for rod differentiation. Nrl knockout mice fail to form rods, generating all cone or S-cone-like (SCL) photoreceptors in the retina, whereas ectopic expression of Nrl using a cone-rod homeobox (Crx) promoter (CrxpNrl) forms all rods. Here, we examined rod and cone pre-synapse development, including axonal elongation, terminal shaping, and synaptic lamination in the outer plexiform layer (OPL) in the presence or absence of Nrl. We show that NRL loss and knockdown result in delayed OPL maturation and plasticity with aberrant dendrites of bipolar neurons. The integrated analyses of the transcriptome in developing rods and SCLs with NRL CUT&RUN and synaptic gene ontology analyses identified G protein subunit beta (Gnb) 1 and p21 (RAC1) activated kinase 5 (Pak5 or Pak7) transcripts were upregulated in developing rods and down-regulated in developing SCLs. Notably, Gnb1 and Gnb5 are rod dominant, and Gnb3 is enriched in cones. NRL binds to the genes of Gnb1, Gnb3, and Gnb5. NRL also regulates pre-synapse ribbon genes, and their expression is altered in rods and SCLs. Our study of histological and gene analyses provides new insights into the morphogenesis of photoreceptor pre-synapse development and regulation of associated genes in the developing retina. Full article
(This article belongs to the Special Issue Retinal Diseases: From Molecular Mechanisms to Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Wild-type outer plexiform layer (OPL) development. (<b>A</b>) Developing OPL stained by anti-Ribeye (synaptic ribbons, green) and anti-Calbindin (horizontal neurons, red). Nuclei stained with DAPI. The boxed areas are shown in insets with higher magnification. OPL (arrowheads) and separate fields of dendrites and axon branches from horizontal neurons (arrows) are shown. (<b>B</b>) Developing OPL stained by anti-Ribeye (green) and anti-PKCα (rod bipolar neurons, red). Clusters of pedicle ribbons (arrows) are shown. (<b>C</b>) Developing rod photoreceptors. Three-dimensional Volocity converted confocal images labeled by in vivo electroporation of <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span> plasmids. Insert at P6 is shown with higher magnification. (<b>D</b>) Growth cone-like structure at P6 terminals of rod photoreceptors. Confocal images displayed at z-thickness of 0.5 μm from synaptic terminal to axon stalk, labeled by in vivo electroporation of <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>. (<b>E</b>) Schematic summary of OPL and photoreceptor synapse development. Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; OPL, outer plexiform layer; DAPI, 4′,6-diamidino-2-phenylindole; P, postnatal day; PKCα, protein kinase C alpha. Scale bars, 10 μm in (<b>A</b>,<b>B</b>) and 5 μm in (<b>D</b>).</p>
Full article ">Figure 2
<p>Wild-type spherule and pedicle. (<b>A</b>) P14 vertical retina sections stained by anti-Reep6 (spherules; green) and PNA (pedicles; red) (upper panel) or anti-CAR (M-cone, green) and PNA (S-cone, red) (lower panel). (<b>B</b>) Horizontal OPL images of retina whole mounts stained by anti-CAR and PNA (upper panels). Pure M-cone (arrows) and S-cone (arrowheads) pedicles are observed. The graph shows the distribution (%, average ± SEM) of CAR pedicle areas in OPL of P14, 21, and 28 whole mount retinas. Over 180 CAR positive pedicles were measured from 3 wild-type C57BL/6J retinas. The area distribution after 30 μm<sup>2</sup> is magnified on the left side. (<b>C</b>) Spherules (green) and a pedicle (red) in CD1 retina whole mount labeled by in vivo electroporation of <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span> and <span class="html-italic">S-opsinp-tdT</span>. Telodendria (yellow arrows) are observed. (<b>D</b>) Representative images of M/S-, S-cone pedicles and spherules and their area size comparison. Telodendria (yellow arrows) are observed in cones. M-cone and pure S-cone pedicles are segregated by anti-M-opsin staining in the retina whole mounts labeled by <span class="html-italic">S-opsin</span>p-<span class="html-italic">tdT</span> electroporation. M/S-pedicles (n = 54), S-pedicles (n = 14), and Rod spherules (n = 64) from 3 to 5 wild-type CD1 retinas were measured. The graph displays the average ± SD of each: 70.79 ± 21.48 μm<sup>2</sup> for M pedicles, 45.91 ± 18.97 μm<sup>2</sup> for S pedicles, and 2.64 ± 0.81 μm<sup>2</sup> for rod spherules. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. Abbreviations: CAR, cone arrestin; PNA, peanut agglutin lectin; <span class="html-italic">S-opsin</span> promoter-driven <span class="html-italic">tdTomato (S-opsinp-tdT</span>); P, postnatal day; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer. Scale bars, 10 μm in (<b>A</b>)<b>,</b> and 5 μm in (<b>B</b>–<b>D</b>).</p>
Full article ">Figure 3
<p>Photoreceptor pre-synapse terminals in <span class="html-italic">Nrl</span><sup>-/-</sup> retina. (<b>A</b>) Vertical retina sections of P18 <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> and <span class="html-italic">Nrl</span><sup>-/-</sup>/<span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> mice, stained by anti-Ribeye (red). (<b>B</b>) Horizontal OPL of retina whole mounts in P18 <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> and <span class="html-italic">Nrl</span><sup>-/-</sup>/<span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> mice, stained by anti-Ribeye (red). (<b>C</b>) Horizontal OPL of retina whole mounts in P14 and P28 <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> and <span class="html-italic">Nrl</span><sup>-/-</sup>/<span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> mice, stained by anti-CAR (red). Arrows indicate pre-synaptic terminals in small size. Abbreviations: <span class="html-italic">Nrl</span>, neural retina leucine zipper; <span class="html-italic">GFP</span>, green fluorescent protein; <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span>, <span class="html-italic">Nrl</span> promoter-driven <span class="html-italic">GFP</span>; P, postnatal day; CAR, cone arrestin. Scale bars, 1 μm in magnified box of (<b>A</b>), 5 μm in (<b>A</b>,<b>C</b>), and 2.5 μm in (<b>B</b>).</p>
Full article ">Figure 4
<p>Pre-synapse comparison in wild-type rod, M/S-cone, pure S-cone, and <span class="html-italic">Nrl</span><sup>-/-</sup> or <span class="html-italic">Nrl</span> knockdown S-cone-like (SCL) photoreceptors. (<b>A</b>) Representative Volocity 3D images of wild-type rod, M/S-cone, pure S-cone, and <span class="html-italic">Nrl</span><sup>-/-</sup> SCL photoreceptors, taken from wild-type or <span class="html-italic">Nrl</span><sup>-/-</sup> retina whole mounts labeled by <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>, <span class="html-italic">S-opsinp-tdT</span>. Rods (green only), cones (red only) and SCLs (mixed green and red) were imaged. M/S- and pure S-cones were differentiated by staining with an anti-M-opsin antibody. (<b>B</b>) Representative confocal images of pre-synapse terminals of wild-type rod, M/S-cone, pure S-cone, and <span class="html-italic">Nrl</span><sup>-/-</sup> SCL photoreceptors. (<b>C</b>) Size distribution of pre-synapses in wild-type rod (n = 64), M/S-cone (n = 54), pure S-cone (n = 14) and <span class="html-italic">Nrl</span><sup>-/-</sup> SCL (P14, n = 25; P21, n = 38) photoreceptors. (<b>D</b>) Representative Volocity 3D images of P28 retina whole mounts labeled by electroporation of scrambled or <span class="html-italic">Nrl</span> shRNA plasmid (sh<span class="html-italic">Nrl</span>) with <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span> (2:1 ratio). (<b>E</b>) Representative confocal images of pre-synaptic terminals expressing scrambled or <span class="html-italic">Nrl</span> shRNA. (<b>F</b>) Size distribution of pre-synapses in control (P14, n = 248; P21, n = 64; P28, n = 98) and developing <span class="html-italic">Nrl</span> shRNA SCL photoreceptors (P14, n = 246; P21, n = 31; P28, n = 124; P35, n = 36) labeled with <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>. Data of area in measurement were analyzed by one-way ANOVA (Tukey or Kriskal–Wallis test) and T-test (two-tailed) in Prism. *, statistically meaningful in one-way ANOVA and T-test; *t, statistically meaningful in T-test. Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; 3D, three-dimensional; <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>, <span class="html-italic">Nrl</span> promoter-driven enhanced <span class="html-italic">GFP</span>; <span class="html-italic">S-opsin</span> promoter-driven <span class="html-italic">tdTomato</span> (<span class="html-italic">S-opsinp-tdT</span>); P, postnatal day; SCL, S-cone-like; shRNA, short hairpin ribonucleic acid. Scale bars, 5 μm in (<b>B</b>,<b>E</b>).</p>
Full article ">Figure 5
<p>Outer plexiform layer development and synaptic connection in <span class="html-italic">Nrl</span><sup>-/-</sup> retina. (<b>A</b>) Developing (P10 to P17) retinas of wild-type and <span class="html-italic">Nrl</span><sup>-/-</sup> mice stained by anti-Ribeye (green) and anti-PKCα (red). Clustered pedicle ribbons (white dotted lines) and dendritic tips of rod bipolar neurons without synaptic ribbons (yellow arrows) are observed. (<b>B</b>) Comparison of OPL thickness in developing wild-type and <span class="html-italic">Nrl</span><sup>-/-</sup> retinas. Measurement was quantified on five images of the middle retina (with optic nerve head) from each of three to four animals in different developing stages. Values represent mean ± SD. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. (<b>C</b>) Comparison of the ribbon distribution in OPL. Distance of ribbon location from the ONL bottom when the OPL thickness is considered 1.0. The location of individual ribbons was measured with each OPL thickness in over two images from each of three to four animals. Values represent mean ± SD. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. (<b>D</b>) Number comparison (%) of rod bipolar neuron dendritic tips with or without ribbons aligned at their tops. Dendritic tips of rod bipolar neurons were measured at P10 (<span class="html-italic">WT</span>, n = 363; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 627), P12 (<span class="html-italic">WT</span>, n = 445; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 953), P14 (<span class="html-italic">WT</span>, n = 433; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 691), and P17 (<span class="html-italic">WT</span>, n = 197; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 269). Values represent mean ± SD. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; P, postnatal day; PKCα, Protein Kinase C alpha; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer. Scale bars, 5 μm in (<b>A</b>).</p>
Full article ">Figure 6
<p>Synaptic plasticity in <span class="html-italic">Nrl</span> transgenic retinas. (<b>A</b>) 1.5-month retinas of wild-type, <span class="html-italic">Nrl</span><sup>-/-</sup> (cone-only) and <span class="html-italic">Crx</span>p<span class="html-italic">Nrl</span> (rod-only) mice stained by anti-PKCα (rod bipolar neurons, green) and anti-Goα (all ON bipolar neurons, red). (<b>B</b>) Retinas from 1.5-month-old <span class="html-italic">Clm-GFP</span> (type 9 cone bipolar neurons, green), <span class="html-italic">Clm-GFP</span>/<span class="html-italic">Nrl</span><sup>-/-</sup>, and <span class="html-italic">Clm-GFP</span>/<span class="html-italic">Crx</span>p<span class="html-italic">Nrl</span> mice stained with anti-Ribeye (red). Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; <span class="html-italic">Crx</span>p<span class="html-italic">Nrl</span>, <span class="html-italic">Cone-rod homeobox</span> promoter-driven <span class="html-italic">Nrl</span>; Clm, clomeleon; GFP, green fluorescent protein; PKCα, Protein Kinase C alpha; Goα, guanine nucleotide-binding protein alpha subunit; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer. Scale bars, 10 μm in (<b>A</b>,<b>B</b>).</p>
Full article ">Figure 7
<p>RNA-seq and CUT&amp;RUN-seq analyses of photoreceptor pre-synapse genes. (<b>A</b>) Schematic drawing of NRL gene regulation in rod photoreceptors. NRL activates rod genes and suppresses cone genes. (<b>B</b>,<b>C</b>) Venn diagrams displaying differently expressed genes in rods, SCLs, and NRL-binding genes. (<b>D</b>–<b>F</b>) SynGO visualizations of upRod-NRL, upRod-downSCL, and upSCL-NRL. (<b>G</b>) SynGO synapse genes, in order from P2 to P28 in transcript per million (TPM) heatmaps: SynGO upRod-NRL (left), SynGO upRod-downSCL (upper right), SynGO upSCL-NRL (down right). Genes in which transcripts are upregulated in developing rods and down-regulated in developing SCLs up to P28 are indicated with a green asterisk, and genes in which transcripts are down-regulated in rods and upregulated in developing SCLs are indicated with a purple asterisk. Known ribbon-associated genes are indicated with a yellow dot. (<b>H</b>) <span class="html-italic">Gnb1</span>, <span class="html-italic">Gnb3</span>, and <span class="html-italic">Gnb5</span> expression in developing rods and SCLs measured by RNAseq dataset (TPM). The line plots (average ± SD) from all relevant transcripts in 2 to 4 bio-replicates were generated using ggplot2 from R studio. (<b>I</b>) Ribbon synapse genes, in order from P2 to P29 in the TPM heatmap. Gene names in magenta correspond to genes showing higher expression in SCLs than rods at P28. Green gene names correspond to genes showing higher expression in rods than SCLs at P28. Gene names in bold font indicate that NRL binds to the genes. Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; upRod-NRL, upregulated NRL binding genes in developing WT rods; upSCL-NRL, upregulated NRL binding genes in developing S-cone-like photoreceptors; downSCL, down-regulated genes in developing S-cone-like photoreceptors; adj. <span class="html-italic">p</span>, adjusted <span class="html-italic">p</span>-value; TPM, transcript per million; SynGO, synaptic Gene Ontologies and annotations.</p>
Full article ">
20 pages, 3262 KiB  
Article
Investigating the Role of Cannabinoid Type 1 Receptors in Vascular Function and Remodeling in a Hypercholesterolemic Mouse Model with Low-Density Lipoprotein–Cannabinoid Type 1 Receptor Double Knockout Animals
by Zsolt Vass, Kinga Shenker-Horváth, Bálint Bányai, Kinga Nóra Vető, Viktória Török, Janka Borbála Gém, György L. Nádasy, Kinga Bernadett Kovács, Eszter Mária Horváth, Zoltán Jakus, László Hunyady, Mária Szekeres and Gabriella Dörnyei
Int. J. Mol. Sci. 2024, 25(17), 9537; https://doi.org/10.3390/ijms25179537 - 2 Sep 2024
Viewed by 734
Abstract
Hypercholesterolemia forms the background of several cardiovascular pathologies. LDL receptor-knockout (LDLR-KO) mice kept on a high-fat diet (HFD) develop high cholesterol levels and atherosclerosis (AS). Cannabinoid type 1 receptors (CB1Rs) induce vasodilation, although their role in cardiovascular pathologies is still controversial. [...] Read more.
Hypercholesterolemia forms the background of several cardiovascular pathologies. LDL receptor-knockout (LDLR-KO) mice kept on a high-fat diet (HFD) develop high cholesterol levels and atherosclerosis (AS). Cannabinoid type 1 receptors (CB1Rs) induce vasodilation, although their role in cardiovascular pathologies is still controversial. We aimed to reveal the effects of CB1Rs on vascular function and remodeling in hypercholesterolemic AS-prone LDLR-KO mice. Experiments were performed on a newly established LDLR and CB1R double-knockout (KO) mouse model, in which KO and wild-type (WT) mice were kept on an HFD or a control diet (CD) for 5 months. The vascular functions of abdominal aorta rings were tested with wire myography. The vasorelaxation effects of acetylcholine (Ach, 1 nM–1 µM) were obtained after phenylephrine precontraction, which was repeated with inhibitors of nitric oxide synthase (NOS) and cyclooxygenase (COX), Nω-nitro-L-arginine (LNA), and indomethacin (INDO), respectively. Blood pressure was measured with the tail-cuff method. Immunostaining of endothelial NOS (eNOS) was carried out. An HFD significantly elevated the cholesterol levels in the LDLR-KO mice more than in the corresponding WT mice (mean values: 1039 ± 162 mg/dL vs. 91 ± 18 mg/dL), and they were not influenced by the presence of the CB1R gene. However, with the defect of the CB1R gene, damage to the Ach relaxation ability was moderated. The blood pressure was higher in the LDLR-KO mice compared to their WT counterparts (systolic/diastolic values: 110/84 ± 5.8/6.8 vs. 102/80 ± 3.3/2.5 mmHg), which was significantly elevated with an HFD (118/96 ± 1.9/2 vs. 100/77 ± 3.4/3.1 mmHg, p < 0.05) but attenuated in the CB1R-KO HFD mice. The expression of eNOS was depressed in the HFD WT mice compared to those on the CD, but it was augmented if CB1R was knocked out. This newly established double-knockout mouse model provides a tool for studying the involvement of CB1Rs in the development of hypercholesterolemia and atherosclerosis. Our results indicate that knocking out the CB1R gene significantly attenuates vascular damage in hypercholesterolemic mice. Full article
(This article belongs to the Special Issue Advances in Animal Models in Biomedical Research, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Body weight values of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control diet or a high-fat diet (one-way ANOVA with pairwise comparisons and Bonferroni post hoc test) (***, <span class="html-italic">p</span> &lt; 0.001; ###, <span class="html-italic">p</span> &lt; 0.001; <span class="html-italic">n</span> = 5–10). Mean ± SEM values are indicated here, with dots showing individual data points. Abbreviations: CD, control diet; HFD, high-fat diet; CB<sub>1</sub>R+/+, CB<sub>1</sub>R wild type; CB<sub>1</sub>R−/−, CB<sub>1</sub>R knockout; LDLR+/+, low-density lipoprotein receptor wild type; LDLR−/−, low-density lipoprotein receptor knockout.</p>
Full article ">Figure 2
<p>Cholesterol levels of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control diet or a high-fat diet (one-way ANOVA with Holm–Sidak comparisons and two-way ANOVA with Bonferroni post hoc test) (**, <span class="html-italic">p</span> = 0.006 and ***, <span class="html-italic">p</span> &lt; 0.001 between CD and HFD groups) (#, <span class="html-italic">p</span> = 0.013; ##, <span class="html-italic">p</span> = 0.002; and ###, <span class="html-italic">p</span> &lt; 0.001 between LDLR+/+ and LDLR−/− groups) (<span class="html-italic">n</span> = 3–5). Mean ± SEM values are indicated here, with dots showing individual data points. Abbreviations: CD, control diet; HFD, high-fat diet; CB<sub>1</sub>R, cannabinoid type 1 receptor; CB<sub>1</sub>R+/+, endocannabinoid type 1 receptor wild type; CB<sub>1</sub>R−/−, endocannabinoid type 1 knockout; LDLR, low-density lipoprotein receptor; LDLR+/+, low-density lipoprotein receptor wild type; LDLR−/−, low-density lipoprotein receptor knockout.</p>
Full article ">Figure 3
<p>Systolic and diastolic blood pressures of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control diet or a high-fat diet. Panel (<b>A</b>): Systolic blood pressure of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control diet or a high-fat diet (<span class="html-italic">n</span> = 5–10). Panel (<b>B</b>): Diastolic blood pressure of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control diet or a high-fat diet (<span class="html-italic">n</span> = 5–10). +++, <span class="html-italic">p</span> &lt; 0.001 between CB<sub>1</sub>R+/+ and CB<sub>1</sub>R−/−; ###, <span class="html-italic">p</span> &lt; 0.001 between LDLR+/+ and LDLR−/− (one-way ANOVA with Holm–Sidak pairwise comparisons, <span class="html-italic">n</span> = 5–10). Mean ± SEM values are indicated here, with dots showing individual data points. Abbreviations: CD, control diet; HFD, high-fat diet; CB<sub>1</sub>R+/+, endocannabinoid type 1 receptor wild type; CB<sub>1</sub>R−/−, endocannabinoid type 1 knockout; LDLR+/+, low-density lipoprotein receptor wild type; LDLR−/−, low-density lipoprotein receptor knockout.</p>
Full article ">Figure 4
<p>Acetylcholine-induced endothelium-dependent vasodilation in aortic segments of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control or a high-fat diet. Panel (<b>A</b>): Dose–response relaxation curves in relation to Ach in CB<sub>1</sub>R-wild-type groups with different LDLR genotypes and diets (##, <span class="html-italic">p</span> = 0.008; one-way ANOVA with Bonferroni post hoc test; <span class="html-italic">n</span> = 5–10). Panel (<b>B</b>): Dose–response relaxation curves in relation to Ach in CB<sub>1</sub>R-knockout groups with different LDLR genotypes and diets (#, <span class="html-italic">p</span> = 0.041; two-way ANOVA with Holm–Sidak post hoc test; <span class="html-italic">n</span> = 6–9). Panel (<b>C</b>): Dose–response relaxation curves in relation to Ach in control-diet groups with different LDLR and CB<sub>1</sub>R genotypes (#, <span class="html-italic">p</span> = 0.047; two-way ANOVA with Holm–Sidak post hoc test; <span class="html-italic">n</span> = 5–7). Panel (<b>D</b>): Dose–response relaxation curves in relation to Ach in HFD groups (<span class="html-italic">n</span> = 5–10) with different LDLR and CB<sub>1</sub>R genotypes (#, <span class="html-italic">p</span> = 0.043; one-way ANOVA with Bonferroni post hoc test). Data are shown as mean ± SEM values. Relaxation data were calculated as percent values of the precontraction level. Abbreviations: CD, control diet; HFD, high-fat diet; CB<sub>1</sub>R+/+, endocannabinoid type 1 receptor wild type; CB<sub>1</sub>R−/−, endocannabinoid type 1 receptor knockout; LDLR+/+, low-density lipoprotein receptor wild type; LDLR−/−, low-density lipoprotein receptor knockout.</p>
Full article ">Figure 5
<p>Effects of specific inhibitors (Nω-nitro-L-arginine and indomethacin, inhibitors of NOS and COX, respectively) on acetylcholine-induced relaxation responses in aortas of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control diet or a high-fat diet. Panel (<b>A</b>): Effects of inhibitors on acetylcholine-induced vasodilation in CB<sub>1</sub>R+/+, LDLR+/+, control-diet mice (<span class="html-italic">n</span> = 5–6). Panel (<b>B</b>): Effects of inhibitors on acetylcholine-induced vasodilation in the CB<sub>1</sub>R−/−, LDLR+/+, control-diet group (<span class="html-italic">n</span> = 6). Panel (<b>C</b>): Effects of inhibitors on acetylcholine-induced vasodilation in CB<sub>1</sub>R+/+, LDLR−/−, control-diet group (<span class="html-italic">n</span> = 7). Panel (<b>D</b>): Effects of inhibitors on acetylcholine-induced vasodilation in the CB<sub>1</sub>R−/−, LDLR−/−, control-diet group (<span class="html-italic">n</span> = 6). Panel (<b>E</b>): Effects of inhibitors on acetylcholine-induced vasodilation in CB<sub>1</sub>R+/+, LDLR+/+, high-fat-diet mice (<span class="html-italic">n</span> = 9–10). Panel (<b>F</b>): Effects of inhibitors on acetylcholine-induced vasodilation in CB<sub>1</sub>R−/−, LDLR+/+, high-fat-diet mice (<span class="html-italic">n</span> = 9). Panel (<b>G</b>): Effects of inhibitors on acetylcholine-induced vasodilation in CB<sub>1</sub>R+/+, LDLR−/−, high-fat-diet mice (<span class="html-italic">n</span> = 5). Panel (<b>H</b>): Effects of inhibitors on acetylcholine-induced vasodilation in the CB<sub>1</sub>R−/−, LDLR−/−, high-fat-diet group (<span class="html-italic">n</span> = 7). Data are shown as mean ± SEM values. <span class="html-italic">p</span> &lt; 0.05 values were considered significant. *, <span class="html-italic">p</span> &lt; 0.05 and ***, <span class="html-italic">p</span> &lt; 0.001 between vehicle- and LNA-treated groups; #, <span class="html-italic">p</span> &lt; 0.05 and ##, <span class="html-italic">p</span> &lt; 0.01 between vehicle- and INDO-treated groups. Statistics were calculated via a one-way ANOVA with Holm–Sidak comparisons or a two-way ANOVA with the Bonferroni post hoc test. Ranking was made with Kruskal–Wallis and Dunn’s tests. Abbreviations: CD, control diet; HFD, high-fat diet; INDO, indomethacin; LNA, Nω-nitro-L-arginine; CB<sub>1</sub>R+/+, cannabinoid type 1 receptor wild type; CB<sub>1</sub>R−/−, cannabinoid type 1 receptor knockout; LDLR+/+, LDL receptor wild type; LDLR−/−, LDL receptor knockout. Relaxation data were calculated as percent values of the precontraction level.</p>
Full article ">Figure 6
<p>Effects of the specific inhibitor Nω-nitro-L-arginine on Ach-induced relaxation in aortas of mice with wild-type/knocked-out CB<sub>1</sub>R and LDLR kept on a control or a high-fat diet, normalized to control values. Attenuation of Ach-induced relaxation with LNA is shown in percent values. Panel (<b>A</b>): Attenuation of Ach-induced relaxation with LNA in CB<sub>1</sub>R+/+, LDLR+/+, CD and HFD groups, <span class="html-italic">n</span> = 5–10. Panel (<b>B</b>): Attenuation of Ach-induced relaxation with LNA in CB<sub>1</sub>R+/+, LDLR−/−, CD and HFD groups, <span class="html-italic">n</span> = 5–7. Panel (<b>C</b>): Attenuation of Ach-induced relaxation with LNA in CB<sub>1</sub>R−/−, LDLR+/+, CD and HFD groups, <span class="html-italic">n</span> = 6–9. Panel (<b>D</b>): Attenuation of Ach-induced relaxation with LNA in CB<sub>1</sub>R−/−, LDLR−/−, CD and HFD groups, <span class="html-italic">n</span> = 6–7. <span class="html-italic">p</span>-values &lt; 0.05 were considered significant. *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001 between CD and HFD groups in the same genotype (one-way ANOVA with Bonferroni post hoc test). Mean ± SEM values are indicated here, with dots showing individual data points. Abbreviations: CD, control diet; HFD, high-fat diet; LNA, Nω-nitro-L-arginine; CB<sub>1</sub>R+/+, cannabinoid type 1 receptor wild type; CB<sub>1</sub>R−/−, cannabinoid type 1 receptor knockout; LDLR+/+, LDL receptor wild type; LDLR−/−, LDL receptor knockout. Relaxation data were calculated as percent values of the precontraction level.</p>
Full article ">Figure 7
<p>Expression of endothelial nitric oxide synthase (eNOS) in abdominal aortas of LDLR+/+ mice with wild-type or knocked-out CB<sub>1</sub>R kept on a control diet or a high-fat diet, <span class="html-italic">n</span> = 4–7. **, <span class="html-italic">p</span> = 0.016 between control-diet (CD) and high-fat-diet (HFD) groups in the same genotype. Mean ± SEM values are indicated here, with dots showing individual data points. Abbreviations: CD, control diet; HFD, high-fat diet; OD, optical density; CB<sub>1</sub>R+/+, cannabinoid type 1 receptor wild type; CB<sub>1</sub>R−/−, cannabinoid type 1 receptor knockout; LDLR+/+, LDL receptor wild type. Panel (<b>A</b>): Optical density (OD) levels of eNOS expression. Panel (<b>B</b>): Representative photos indicating eNOS expression chosen form 4–7 slides of each group.</p>
Full article ">
27 pages, 6735 KiB  
Article
Novel Role of the ALPI Gene Associated with Constipation Caused by Complement Component 3 Deficiency
by Hee Jin Song, Ji Eun Kim, Yu Jeong Roh, Ayun Seol, Tae Ryeol Kim, Ki Ho Park, Eun Seo Park, Jin Tae Hong, Sun Il Choi and Dae Youn Hwang
Int. J. Mol. Sci. 2024, 25(17), 9530; https://doi.org/10.3390/ijms25179530 - 2 Sep 2024
Viewed by 328
Abstract
Complement component 3 (C3) deficiency has recently been reported as one of the novel causes of constipation. To identify a unique gene specific to constipation caused by C3 deficiency, the total RNA extracted from the mid colon of C3 knockout (C3 KO) mice [...] Read more.
Complement component 3 (C3) deficiency has recently been reported as one of the novel causes of constipation. To identify a unique gene specific to constipation caused by C3 deficiency, the total RNA extracted from the mid colon of C3 knockout (C3 KO) mice was hybridized to oligonucleotide microarrays, and the function of the candidate gene was verified in in vitro and in vivo models. C3 KO mice used for microarrays showed definite phenotypes of constipation. Overall, compared to the wild type (WT), 1237 genes were upregulated, and 1292 genes were downregulated in the C3 KO mice. Of these, the major genes included were lysine (K)-specific demethylase 5D (KDM5D), olfactory receptor 870 (Olfr870), pancreatic lipase (PNLIP), and alkaline phosphatase intestinal (ALPI). Specifically, the ALPI gene was selected as a novel gene candidate based on alterations during loperamide (Lop)-induced constipation and intestinal bowel disease (IBD). The upregulation of ALPI expression treated with acetate recovered the expression level of mucin-related genes in primary epithelial cells of C3 KO mice as well as most phenotypes of constipation in C3 KO mice. These results indicate that ALPI plays an important role as the novel gene associated with C3 deficiency-induced constipation. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Stool excretion, GI transit ratio and mid colon histology of C3 KO mice. (<b>A</b>) Stools excretion. Digital camera images of the stools were taken immediately after collection from the metabolic cage. The total number and water contents of stools were measured as described in the Materials and Methods. Five to six mice per group were used for the stool sample collection, and each parameter was assayed in duplicates. (<b>B</b>) GI transit ratio and intestine length. The total intestinal tract was excised from mice of the subset groups treated with charcoal meal powder. The arrows indicate the position of the charcoal meal. Five to six mice per group were used in the GI transit ratio test, and the charcoal meal transit distance, total intestine length and colon length were measured in duplicates. (<b>C</b>) Histological structures of the mid colons. The H&amp;E-stained sections of the mid colon from the WT and KO mice were observed at 100× magnification using a light microscope. Five to six mice per group were used in the histological analysis, and each parameter was measured in duplicates in two different slides. The data were presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. WT mice. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; GI, gastrointestinal; H&amp;E, hematoxylin and eosin.</p>
Full article ">Figure 1 Cont.
<p>Stool excretion, GI transit ratio and mid colon histology of C3 KO mice. (<b>A</b>) Stools excretion. Digital camera images of the stools were taken immediately after collection from the metabolic cage. The total number and water contents of stools were measured as described in the Materials and Methods. Five to six mice per group were used for the stool sample collection, and each parameter was assayed in duplicates. (<b>B</b>) GI transit ratio and intestine length. The total intestinal tract was excised from mice of the subset groups treated with charcoal meal powder. The arrows indicate the position of the charcoal meal. Five to six mice per group were used in the GI transit ratio test, and the charcoal meal transit distance, total intestine length and colon length were measured in duplicates. (<b>C</b>) Histological structures of the mid colons. The H&amp;E-stained sections of the mid colon from the WT and KO mice were observed at 100× magnification using a light microscope. Five to six mice per group were used in the histological analysis, and each parameter was measured in duplicates in two different slides. The data were presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. WT mice. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; GI, gastrointestinal; H&amp;E, hematoxylin and eosin.</p>
Full article ">Figure 2
<p>Volume and scatter plots of the normalized microarray in C3 KO mice. (<b>A</b>) Volume plot of the WT mice and C3 KO mice. (<b>B</b>) Scatter plot of the WT and C3 KO mice. (<b>C</b>) Differentially regulated probe count. The number of genes expressed differently between WT and C3 KO mice were represented as 1.5 FC and 2 FC. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; FC, fold change.</p>
Full article ">Figure 3
<p>Transcription level of the upregulated genes and downregulated genes. Transcription level of <span class="html-italic">EDN1</span> (<b>A</b>) and <span class="html-italic">NXPE4</span> (<b>B</b>). Transcription level of <span class="html-italic">RETNLB</span> (<b>C</b>) and <span class="html-italic">ALPI</span> (<b>D</b>). The transcription levels of four genes were measured in the total mRNA of the colon by RT-qPCR using specific primers. The mRNA levels of the four genes were calculated based on the intensity of β-actin as an endogenous control. The preparation of total RNAs was performed on three to five mice per group, RT-qPCR analyses were assayed twice for each total RNA. The data were presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. WT mice. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; <span class="html-italic">EDN1</span>, endothelin-1; <span class="html-italic">NXPE4</span>, neurexophilin and PC-esterase domain family member 4; <span class="html-italic">RETNLB</span>, resistin-like beta; <span class="html-italic">ALPI</span>, alkaline phosphatase intestinal; RT-qPCR, quantitative real-time PCR.</p>
Full article ">Figure 4
<p>KEGG pathway of functional analyses about differential genes between WT and C3 KO mice. (<b>A</b>) Top ten terms with statistical significance in the KEGG pathway. (<b>B</b>) Neuroactive ligand–receptor Interaction. (<b>C</b>) Complement and coagulation cascades. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; KEGG, Kyoto Encyclopedia of Genes and Genomes.</p>
Full article ">Figure 5
<p>Transcription level of <span class="html-italic">C-kit, ALPI</span> and mucin secretion genes in epithelial cells from intestine of C3 KO mice after acetate treatment. (<b>A</b>) Transcription level of <span class="html-italic">C-kit</span> gene in isolated epithelial cells and muscle cells. * <span class="html-italic">p</span> &lt; 0.05 vs. Muscle cells. (<b>B</b>) Transcription level of the <span class="html-italic">ALPI</span> gene in acetate-treated C3 KO epithelial cells. (<b>C</b>–<b>G</b>) Transcription level of <span class="html-italic">MUC1</span>, <span class="html-italic">MUC2</span>, <span class="html-italic">KLF4</span>, <span class="html-italic">AQP3</span> and <span class="html-italic">AQP8</span> in acetate-treated C3 KO epithelial cells. The transcription levels of each gene were measured in the total mRNA of the epithelial cells by RT-qPCR using specific primers. The mRNA levels of each gene were calculated based on the intensity of β-actin as an endogenous control. The preparation of total RNAs was performed on three to five wells per group, RT-qPCR analyses were assayed twice for each total RNA. The data were presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. no treated group. # <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated C3 KO group. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; <span class="html-italic">MUC</span>, mucin; <span class="html-italic">AQP</span>, aquaporin; <span class="html-italic">ALPI</span>, alkaline phosphatase intestinal; C-kit, receptor protein kinase; PBS, phosphate buffer saline; RT-qPCR; quantitative real-time PCR.</p>
Full article ">Figure 6
<p>Transcription and expression levels of the <span class="html-italic">ALPI</span> gene and proteins. (<b>A</b>) Transcription level of the <span class="html-italic">ALPI</span> gene in acetate-treated C3 KO mice. The transcription levels of the <span class="html-italic">ALPI</span> gene were measured in the total mRNA of the epithelial cells by RT-qPCR using specific primers. The mRNA levels of each gene were calculated based on the intensity of β-actin as an endogenous control. The preparation of total RNAs was performed on three to five mice per group, RT-qPCR analyses were assayed twice for each total RNA. (<b>B</b>) Expression levels of ALPI proteins in acetate-treated C3 KO mice. After collecting the mid colons from the C3 KO mice, the expression levels of ALPI proteins were assessed in tissue homogenates using the specific primary antibody and densitometry. The tissue homogenates were prepared from four to six mice per group and the Western blot was analyzed twice for each sample. After normalizing with β-actin as endogenous control, the relative level of ALPI expression was presented as a relative value to the vehicle-treated WT group (<b>C</b>). The data were presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. no treated group. # <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated C3 KO group. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; <span class="html-italic">ALPI</span>, alkaline phosphatase intestinal; RT-qPCR, quantitative real-time PCR.</p>
Full article ">Figure 7
<p>Stool parameters, GI transit and histological structure of colon in acetate-treated C3 KO mice. (<b>A</b>) Stool parameters. The preparation of the total stools was performed on three to five mice per group and each parameter was analyzed twice for each sample. (<b>B</b>) GI transit and total length of GI tract. After collection from the stomach to the anus, the entire GI tract was arranged in a row, and the location of the charcoal meal was marked with arrows. Transit ratio of charcoal meal and total length of GI tract were measured as described in materials and method. The preparation of the GI tract was performed on three to five mice per group, and each parameter was analyzed twice for each mouse. (<b>C</b>) Histological structure of mid colon. The H&amp;E-stained tissue sections were prepared from tissue samples from three to five mice per group, and the pathological factors were analyzed twice for each stained tissue. The data are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. no treated group. # <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated C3 KO group. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; <span class="html-italic">ALPI</span>, alkaline phosphatase intestinal; H&amp;E, hematoxylin and eosin.</p>
Full article ">Figure 7 Cont.
<p>Stool parameters, GI transit and histological structure of colon in acetate-treated C3 KO mice. (<b>A</b>) Stool parameters. The preparation of the total stools was performed on three to five mice per group and each parameter was analyzed twice for each sample. (<b>B</b>) GI transit and total length of GI tract. After collection from the stomach to the anus, the entire GI tract was arranged in a row, and the location of the charcoal meal was marked with arrows. Transit ratio of charcoal meal and total length of GI tract were measured as described in materials and method. The preparation of the GI tract was performed on three to five mice per group, and each parameter was analyzed twice for each mouse. (<b>C</b>) Histological structure of mid colon. The H&amp;E-stained tissue sections were prepared from tissue samples from three to five mice per group, and the pathological factors were analyzed twice for each stained tissue. The data are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. no treated group. # <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated C3 KO group. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; <span class="html-italic">ALPI</span>, alkaline phosphatase intestinal; H&amp;E, hematoxylin and eosin.</p>
Full article ">Figure 8
<p>Transcription level of (<b>A</b>) <span class="html-italic">MUC1</span>, (<b>B</b>) <span class="html-italic">MUC2</span>, (<b>C</b>) <span class="html-italic">AQP3</span> and (<b>D</b>) <span class="html-italic">AQP8</span> gene in the colon tissue of acetate-treated C3 KO mice. The transcription levels of four genes were measured in the total mRNA of the colon tissues by RT-qPCR using specific primers. The mRNA levels of each gene were calculated based on the intensity of β-actin as an endogenous control. The preparation of total RNAs was performed on three to five mice per group, RT-qPCR analyses were assayed twice for each total RNA. The data were presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. no treated group. # <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated group. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; MUC, mucin; AQP, aquaporin.</p>
Full article ">Figure 9
<p>Schedule for acetate treatment into C3 KO mice. After allocation of the WT and C3 KO mice into one of three experimental groups, mice of each group were treated with 5 mM acetate or 1× PBS, and subsequentially collected the stools and colon tissue from these mice. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; RT-qPCR, quantitative real-time PCR; PBS, phosphate buffer saline.</p>
Full article ">Figure 10
<p>Scheme for purification of epithelial cells from intestine of C3 KO mice and acetate treatment. After collection of the epithelial cells from the intestines of WT and C3 KO mice, these cells were treated with 5 mM acetate or 1× PBS, and the expression of several genes were evaluated by RT-qPCR analysis using specific primers. Abbreviation: WT, wild type; C3 KO, complement C3 knockout; <span class="html-italic">ALPI</span>, alkaline phosphatase intestinal; C-kit, receptor protein kinase; PBS, phosphate buffer saline; RT-qPCR; quantitative real-time PCR.</p>
Full article ">
20 pages, 3108 KiB  
Article
ACE2 Knockout Mice Are Resistant to High-Fat Diet-Induced Obesity in an Age-Dependent Manner
by Valéria Nunes-Souza, Natalia Alenina, Fatimunnisa Qadri, Valentina Mosienko, Robson Augusto Souza Santos, Michael Bader and Luiza Antas Rabelo
Int. J. Mol. Sci. 2024, 25(17), 9515; https://doi.org/10.3390/ijms25179515 - 1 Sep 2024
Viewed by 448
Abstract
Angiotensin converting enzyme 2 (ACE2) presents pleiotropic actions. It hydrolyzes angiotensin I (AngI) and angiotensin II (AngII) into angiotensin-(1-9) (Ang-(1-9)) and angiotensin-(1-7) (Ang-(1-7)), respectively, as well as participates in tryptophan uptake in the gut and in COVID-19 infection. Our aim was to investigate [...] Read more.
Angiotensin converting enzyme 2 (ACE2) presents pleiotropic actions. It hydrolyzes angiotensin I (AngI) and angiotensin II (AngII) into angiotensin-(1-9) (Ang-(1-9)) and angiotensin-(1-7) (Ang-(1-7)), respectively, as well as participates in tryptophan uptake in the gut and in COVID-19 infection. Our aim was to investigate the metabolic effect of ACE2 deletion in young adults and elderly mice under conditions of high calorie intake. Male C57Bl/6 (WT) and ACE2-deficient (ACE2-/y) mice were analyzed at the age of 6 and 12 months under standard diet (StD) and high-fat diet (HFD). Under StD, ACE2-/y showed lower body weight and fat depots, improved glucose tolerance, enhanced insulin sensitivity, higher adiponectin, and lower leptin levels compared to WT. This difference was even more pronounced after HFD in 6-month-old mice, but, interestingly, it was blunted at the age of 12 months. ACE2-/y presented a decrease in adipocyte diameter and lipolysis, which reflected in the upregulation of lipid metabolism in white adipose tissue through the increased expression of genes involved in lipid regulation. Under HFD, both food intake and total energy expenditure were decreased in 6-month-old ACE2-/y mice, accompanied by an increase in liquid intake, compared to WT mice, fed either StD or HFD. Thus, ACE2-/y mice are less susceptible to HFD-induced obesity in an age-dependent manner, as well as represent an excellent animal model of human lipodystrophy and a tool to investigate new treatments. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Body weight; (<b>B</b>) Total fat; (<b>C</b>) WAT index of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 and 12 months under standard diet (StD, chow) and high-fat diet (HFD); (<b>D</b>) White adipocyte diameter of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 months (Mon) under StD and HFD. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT.</p>
Full article ">Figure 2
<p>(<b>A</b>) Lipolysis in vivo; (<b>B</b>) Lipolysis in vitro; (<b>C</b>) Relative expression of <span class="html-italic">lipoprotein lipase</span> (<span class="html-italic">LPL</span>) in white adipose tissue (WAT); (<b>D</b>) Relative expression of <span class="html-italic">Adrβ3 receptor</span> in WAT; (<b>E</b>) Relative expression of <span class="html-italic">hormone-sensitive lipase</span> (<span class="html-italic">HSL</span>) in WAT; (<b>F</b>) Relative expression of <span class="html-italic">Adiponectin</span> in WAT. (<b>G</b>) Adiponectin levels in plasma; (<b>H</b>) Relative expression of <span class="html-italic">Leptin</span> in WAT; (<b>I</b>) Leptin levels in plasma of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 and 12 months (Mon) under standard diet (StD) and high-fat diet (HFD). Each point on the graph represents the mean ± SEM. ANOVA (two ways): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 KO vs. WT. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT. <sup>###</sup> ACE2<sup>-/y</sup> StD vs. WT StD; <sup>ΦΦ</sup> WT HFD vs. WT StD; <sup>δδδ</sup> ACE2<sup>-/y</sup> HFD vs. WT HFD.</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>) Lipolysis in vivo; (<b>B</b>) Lipolysis in vitro; (<b>C</b>) Relative expression of <span class="html-italic">lipoprotein lipase</span> (<span class="html-italic">LPL</span>) in white adipose tissue (WAT); (<b>D</b>) Relative expression of <span class="html-italic">Adrβ3 receptor</span> in WAT; (<b>E</b>) Relative expression of <span class="html-italic">hormone-sensitive lipase</span> (<span class="html-italic">HSL</span>) in WAT; (<b>F</b>) Relative expression of <span class="html-italic">Adiponectin</span> in WAT. (<b>G</b>) Adiponectin levels in plasma; (<b>H</b>) Relative expression of <span class="html-italic">Leptin</span> in WAT; (<b>I</b>) Leptin levels in plasma of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 and 12 months (Mon) under standard diet (StD) and high-fat diet (HFD). Each point on the graph represents the mean ± SEM. ANOVA (two ways): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 KO vs. WT. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT. <sup>###</sup> ACE2<sup>-/y</sup> StD vs. WT StD; <sup>ΦΦ</sup> WT HFD vs. WT StD; <sup>δδδ</sup> ACE2<sup>-/y</sup> HFD vs. WT HFD.</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>) Lipolysis in vivo; (<b>B</b>) Lipolysis in vitro; (<b>C</b>) Relative expression of <span class="html-italic">lipoprotein lipase</span> (<span class="html-italic">LPL</span>) in white adipose tissue (WAT); (<b>D</b>) Relative expression of <span class="html-italic">Adrβ3 receptor</span> in WAT; (<b>E</b>) Relative expression of <span class="html-italic">hormone-sensitive lipase</span> (<span class="html-italic">HSL</span>) in WAT; (<b>F</b>) Relative expression of <span class="html-italic">Adiponectin</span> in WAT. (<b>G</b>) Adiponectin levels in plasma; (<b>H</b>) Relative expression of <span class="html-italic">Leptin</span> in WAT; (<b>I</b>) Leptin levels in plasma of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 and 12 months (Mon) under standard diet (StD) and high-fat diet (HFD). Each point on the graph represents the mean ± SEM. ANOVA (two ways): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 KO vs. WT. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT. <sup>###</sup> ACE2<sup>-/y</sup> StD vs. WT StD; <sup>ΦΦ</sup> WT HFD vs. WT StD; <sup>δδδ</sup> ACE2<sup>-/y</sup> HFD vs. WT HFD.</p>
Full article ">Figure 3
<p>Glycemic curve and area under curve (AUC) of the glucose tolerance (<b>A</b>,<b>B</b>) and insulin sensitivity (<b>C,D</b>) of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 (<b>A</b>,<b>C</b>) and 12 (<b>B</b>,<b>D</b>) months (Mon) under standard (StD) and high-fat diet (HFD). (<b>E</b>,<b>F</b>) <span class="html-italic">Insulin receptor</span> (<span class="html-italic">IR</span>) mRNA expression in white adipose tissue (WAT) and muscle, respectively, and (<b>G</b>,<b>H</b>) <span class="html-italic">GLUT4</span> mRNA expression in WAT and muscle, respectively, in 6-month-old animals under StD and HFD. Each point on the graph represents the mean ± SEM. ANOVA (two ways): * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; KO vs. WT.</p>
Full article ">Figure 3 Cont.
<p>Glycemic curve and area under curve (AUC) of the glucose tolerance (<b>A</b>,<b>B</b>) and insulin sensitivity (<b>C,D</b>) of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 (<b>A</b>,<b>C</b>) and 12 (<b>B</b>,<b>D</b>) months (Mon) under standard (StD) and high-fat diet (HFD). (<b>E</b>,<b>F</b>) <span class="html-italic">Insulin receptor</span> (<span class="html-italic">IR</span>) mRNA expression in white adipose tissue (WAT) and muscle, respectively, and (<b>G</b>,<b>H</b>) <span class="html-italic">GLUT4</span> mRNA expression in WAT and muscle, respectively, in 6-month-old animals under StD and HFD. Each point on the graph represents the mean ± SEM. ANOVA (two ways): * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; KO vs. WT.</p>
Full article ">Figure 3 Cont.
<p>Glycemic curve and area under curve (AUC) of the glucose tolerance (<b>A</b>,<b>B</b>) and insulin sensitivity (<b>C,D</b>) of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 (<b>A</b>,<b>C</b>) and 12 (<b>B</b>,<b>D</b>) months (Mon) under standard (StD) and high-fat diet (HFD). (<b>E</b>,<b>F</b>) <span class="html-italic">Insulin receptor</span> (<span class="html-italic">IR</span>) mRNA expression in white adipose tissue (WAT) and muscle, respectively, and (<b>G</b>,<b>H</b>) <span class="html-italic">GLUT4</span> mRNA expression in WAT and muscle, respectively, in 6-month-old animals under StD and HFD. Each point on the graph represents the mean ± SEM. ANOVA (two ways): * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 KO vs. WT. Each bar graph represents the mean ± SEM. ANOVA (one way): * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; KO vs. WT.</p>
Full article ">Figure 4
<p>(<b>A</b>) Superoxide Dismutase (SOD) and (<b>B</b>) Catalase (CAT) activities in white adipose tissue (WAT) of C57Bl/6 and ACE2<sup>-/y</sup> mice at the age of 6 months (Mon) under standard diet (StD) and high-fat diet (HFD). Each bar graph represents the mean ± SEM. ANOVA (one way).</p>
Full article ">Figure 5
<p>❶ The 6-month-old ACE2<sup>-/y</sup> mice are protect against HFD-induced obesity. ❷ The animals presented a decrease in body weight (BW), ❸ total fat, white adipose tissue (WAT) index, white adipocyte diameter, and <span class="html-italic">leptin</span> gene in WAT; decrease in lipolysis in vivo, increase in lipolytic (<span class="html-italic">HSL, Adrβ3</span>) and <span class="html-italic">adiponectin</span> genes in WAT. ❹ Systemically, these effects reflected in the increase in glucose tolerance, insulin sensitivity, and adiponectin, as well as a decrease in leptin protein. ❺ In the gastrocnemius muscle, it is observed that there is a decrease in <span class="html-italic">insulin receptor</span> (<span class="html-italic">IR)</span> gene and % muscle, which possibly reflects in ❻ the decrease in total energy expenditure (TEE) and food intake detected in ACE2<sup>-/y</sup> mice. Regarding the liquid intake, ACE2<sup>-/y</sup> mice present an increase, probably induced by the high levels of AngII, since it stimulates thirst. ❼ The deletion of ACE2 at the age of 12 months did not change BW, fat deposition, or plasma glucose homeostasis under HFD, demonstrating a delayed effect of HFD in ACE2<sup>-/y</sup> mice, suggesting that ACE2 in an old age no longer interferes with these parameters as observed in younger ages, and that the mechanism of protection against HFD-induced obesity could be age-dependent. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
15 pages, 4417 KiB  
Article
Aerobic Exercise Protects against Cardiotoxin-Induced Skeletal Muscle Injury in a DDAH1-Dependent Manner
by Fei Feng, Kai Luo, Xinyi Yuan, Ting Lan, Siyu Wang, Xin Xu and Zhongbing Lu
Antioxidants 2024, 13(9), 1069; https://doi.org/10.3390/antiox13091069 - 1 Sep 2024
Viewed by 446
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a critical enzyme that regulates nitric oxide (NO) signaling through the degradation of asymmetric dimethylarginine (ADMA). Previous studies have revealed a link between the beneficial effects of aerobic exercise and the upregulation of DDAH1 in bones and hearts. [...] Read more.
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a critical enzyme that regulates nitric oxide (NO) signaling through the degradation of asymmetric dimethylarginine (ADMA). Previous studies have revealed a link between the beneficial effects of aerobic exercise and the upregulation of DDAH1 in bones and hearts. We previously reported that skeletal muscle DDAH1 plays a protective role in cardiotoxin (CTX)-induced skeletal muscle injury and regeneration. To determine the effects of aerobic exercise on CTX-induced skeletal muscle injury and the role of DDAH1 in this process, wild-type (WT) mice and skeletal muscle-specific Ddah1-knockout (Ddah1MKO) mice were subjected to swimming training for 8 weeks and then injected with CTX. In WT mice, swimming training for 8 weeks significantly promoted skeletal muscle regeneration and attenuated inflammation, oxidative stress, and apoptosis in the gastrocnemius (GA) muscle after CTX injection. These phenomena were associated with increases in the protein expression of PAX7, myogenin, MEF2A, eNOS, SOD2, and peroxiredoxin 5 and decreases in iNOS expression in GA muscles. Swimming training also decreased serum ADMA levels and increased serum nitrate/nitrite (NOx) levels and skeletal muscle DDAH1 expression. Interestingly, swimming training in Ddah1MKO mice had no obvious effect on CTX-induced skeletal muscle injury or regeneration and did not repress the CTX-induced inflammatory response, superoxide generation, or apoptosis. In summary, our data suggest that DDAH1 is important for the protective effect of aerobic exercise on skeletal muscle injury and regeneration. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Aerobic exercise promotes skeletal muscle regeneration in cardiotoxin (CTX)-treated mice. (<b>A</b>) The experimental design of this study is shown in a schematic diagram. On Days 3 and 5 after CTX injection, the body weight (<b>B</b>), the GA weight-to-body weight (GA/BW) ratio (<b>C</b>), and the serum lactate dehydrogenase (LDH) level (<b>D</b>) were measured. (<b>E</b>) Representative cryosections from GA muscle were stained with wheat germ agglutinin (WGA). The myofiber average cross-sectional area (CSA) was measured via ImageJ. The distribution of GA myofiber size in control and CTX-treated mice is shown. Scale bar = 50 μm. (<b>F</b>) Cryosections of the GA muscle were stained with DAPI (blue) and antibodies specific for PAX7 (green) and laminin (red). Scale bar = 20 μm. (<b>G</b>) GA muscle was homogenized in RIPA buffer, and the lysates were examined via Western blotting. In <a href="#antioxidants-13-01069-f001" class="html-fig">Figure 1</a>A–F, N = 5; in <a href="#antioxidants-13-01069-f001" class="html-fig">Figure 1</a>G, N = 3; the values are expressed as the means ± SEMs. ns indicates not significant, * indicates <span class="html-italic">p</span> &lt; 0.05, ** indicates <span class="html-italic">p</span> &lt; 0.01, and *** indicates <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Aerobic exercise attenuated the CTX-induced skeletal muscle inflammatory response. (<b>A</b>) Representative cryosections of GA muscle were stained with hematoxylin and eosin (H&amp;E) and an antibody specific for F4/80 (brown staining). Scale bar = 50 μm. (<b>B</b>) F4/80-positive cell numbers were quantified via ImageJ. (<b>C</b>–<b>I</b>) The mRNA levels of M1 and M2 macrophage-specific markers in each group were measured on Days 3 and 5 after CTX injection. In <a href="#antioxidants-13-01069-f002" class="html-fig">Figure 2</a>A–I, N = 5; the values are expressed as the mean ± SEM. * indicates <span class="html-italic">p</span> &lt; 0.05, ** indicates <span class="html-italic">p</span> &lt; 0.01, and *** indicates <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Aerobic exercise ameliorated CTX-induced apoptosis and oxidative stress in GA muscle. (<b>A</b>) Representative frozen GA muscle sections from each group were stained with DHE, and the relative fluorescence intensity was quantified; scale bar = 50 μm. (<b>B</b>) Representative frozen skeletal muscle sections from each group were subjected to TUNEL, and the number of apoptotic cells was quantified; scale bar = 50 μm. (<b>C</b>) GA muscle lysates were subjected to Western blot analysis. In <a href="#antioxidants-13-01069-f003" class="html-fig">Figure 3</a>A,B, N = 5; in <a href="#antioxidants-13-01069-f003" class="html-fig">Figure 3</a>C, N = 3; the values are expressed as the means ± SEMs. * indicates <span class="html-italic">p</span> &lt; 0.05, ** indicates <span class="html-italic">p</span> &lt; 0.01, and *** indicates <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>DDAH1 in skeletal muscle is involved in the protective effect of aerobic exercise on CTX-induced muscle injury. Serum asymmetric dimethylarginine (ADMA) (<b>A</b>) and nitrate/nitrite (NOx) (<b>B</b>) levels were measured on Days 3 and 5 after CTX injection. (<b>C</b>) DDAH1 expression in the GA muscle was examined via Western blotting. (<b>D</b>) The experimental process for <span class="html-italic">Ddah1</span><sup>MKO</sup> mice is shown in a schematic diagram. On Day 5 after CTX injection, the body weight (<b>E</b>), the GA weight-to-body weight ratio (<b>F</b>) and the serum levels of LDH (<b>G</b>), ADMA (<b>H</b>) and NOx (<b>I</b>) were measured. In <a href="#antioxidants-13-01069-f004" class="html-fig">Figure 4</a>A,B and E–I, N = 5; in <a href="#antioxidants-13-01069-f004" class="html-fig">Figure 4</a>C, N = 3; the values are expressed as the means ± SEMs. * indicates <span class="html-italic">p</span> &lt; 0.05, *** indicates <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Aerobic exercise did not promote CTX-induced skeletal muscle regeneration in <span class="html-italic">Ddah1</span><sup>MKO</sup> mice. (<b>A</b>) Cryosections from the GA muscle in each group were stained with WGA; scale bars = 50 μm. The average myofiber CSA was measured via ImageJ, and the GA myofiber size distributions in control and CTX–treated mice are shown. (<b>B</b>) Cryosections from control and CTX–treated GA muscle were subjected to immunofluorescence, and SCs were labeled with both PAX7 (green) and DAPI (blue). Scale bar = 20 μm. PAX7-positive cells are shown, and each mouse skeletal muscle tissue section was randomly photographed in 10 visual fields, after which statistical analysis was performed. (<b>C</b>) GA muscle was homogenized in RIPA buffer, and the lysates were examined via Western blotting. In <a href="#antioxidants-13-01069-f005" class="html-fig">Figure 5</a>A–E, N = 5; in <a href="#antioxidants-13-01069-f005" class="html-fig">Figure 5</a>F, N = 3; the values are expressed as the means ± SEMs. * indicates <span class="html-italic">p</span> &lt; 0.05, ** indicates <span class="html-italic">p</span> &lt; 0.01, and *** indicates <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Aerobic exercise had no effect on CTX-induced inflammation, oxidative stress or apoptosis in the GA muscle of <span class="html-italic">Ddah1</span><sup>MKO</sup> mice. (<b>A</b>) Representative frozen GA muscle sections were stained with H&amp;E and an antibody specific for F4/80 (brown staining); scale bars = 50 μm. The number of F4/80-positive cells was quantified. (<b>B</b>) The mRNA levels of inflammatory genes were measured via qPCR. (<b>C</b>) Representative frozen GA muscle sections were stained with DHE and TUNEL kits. The relative fluorescence intensity and number of apoptotic cells were quantified, scale bars = 50 μm. (<b>D</b>) GA muscle lysates were examined via Western blotting. In <a href="#antioxidants-13-01069-f006" class="html-fig">Figure 6</a>A–C, N = 5. In <a href="#antioxidants-13-01069-f006" class="html-fig">Figure 6</a>D, N = 3; the values are expressed as the means ± SEMs, * indicates <span class="html-italic">p</span> &lt; 0.05, ** indicates <span class="html-italic">p</span> &lt; 0.01, and *** indicates <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
Back to TopTop