(Go: >> BACK << -|- >> HOME <<)

 
 

Topic Editors

Pathobiology and Extracellular Vesicle Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK
Prof. Dr. Jameel M. Inal
Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, UK

Animal Models of Human Disease 2.0

Abstract submission deadline
closed (30 June 2024)
Manuscript submission deadline
31 August 2024
Viewed by
16293

Topic Information

Dear Colleagues, 

The use of animal models of human disease is critical for furthering our understanding of disease mechanisms, for the discovery of novel targets for treatment, and for translational research. This Topic aims to collect state-of-the-art primary research studies and review articles from international experts and leading groups using animal models to study human diseases. Submissions are welcome on a wide range of animal models and pathologies, including infectious disease, acute injury, regeneration, cancer, autoimmunity, and degenerative and chronic disease.

Prof. Dr. Sigrun Lange
Prof. Dr. Jameel M. Inal
Topic Editors

Keywords

  • animal models
  • human disease
  • pathology
  • pathobiology
  • chronic disease
  • acute injury
  • regeneration
  • infectious disease
  • cancer
  • autoimmunity
  • neurodegenerative disease
  • comparative animal models
  • extracellular vesicles
  • liquid biopsy
  • biomarkers

Participating Journals

Journal Name Impact Factor CiteScore Launched Year First Decision (median) APC
Biomedicines
biomedicines
3.9 5.2 2013 15.3 Days CHF 2600 Submit
Cells
cells
5.1 9.9 2012 17.5 Days CHF 2700 Submit
Current Issues in Molecular Biology
cimb
2.8 2.9 1999 16.8 Days CHF 2200 Submit
Genes
genes
2.8 5.2 2010 16.3 Days CHF 2600 Submit
International Journal of Molecular Sciences
ijms
4.9 8.1 2000 18.1 Days CHF 2900 Submit

Preprints.org is a multidiscipline platform providing preprint service that is dedicated to sharing your research from the start and empowering your research journey.

MDPI Topics is cooperating with Preprints.org and has built a direct connection between MDPI journals and Preprints.org. Authors are encouraged to enjoy the benefits by posting a preprint at Preprints.org prior to publication:

  1. Immediately share your ideas ahead of publication and establish your research priority;
  2. Protect your idea from being stolen with this time-stamped preprint article;
  3. Enhance the exposure and impact of your research;
  4. Receive feedback from your peers in advance;
  5. Have it indexed in Web of Science (Preprint Citation Index), Google Scholar, Crossref, SHARE, PrePubMed, Scilit and Europe PMC.

Published Papers (16 papers)

Order results
Result details
Journals
Select all
Export citation of selected articles as:
19 pages, 4445 KiB  
Article
Simulated Microgravity Alters Gene Regulation Linked to Immunity and Cardiovascular Disease
by Candice G. T. Tahimic, Sonette Steczina, Aimy Sebastian, Nicholas R. Hum, Metadel Abegaz, Masahiro Terada, Maria Cimini, David A. Goukassian, Ann-Sofie Schreurs, Tana M. Hoban-Higgins, Charles A. Fuller, Gabriela G. Loots, Ruth K. Globus and Yasaman Shirazi-Fard
Genes 2024, 15(8), 975; https://doi.org/10.3390/genes15080975 (registering DOI) - 24 Jul 2024
Viewed by 151
Abstract
Microgravity exposure induces a cephalad fluid shift and an overall reduction in physical activity levels which can lead to cardiovascular deconditioning in the absence of countermeasures. Future spaceflight missions will expose crew to extended periods of microgravity among other stressors, the effects of [...] Read more.
Microgravity exposure induces a cephalad fluid shift and an overall reduction in physical activity levels which can lead to cardiovascular deconditioning in the absence of countermeasures. Future spaceflight missions will expose crew to extended periods of microgravity among other stressors, the effects of which on cardiovascular health are not fully known. In this study, we determined cardiac responses to extended microgravity exposure using the rat hindlimb unloading (HU) model. We hypothesized that exposure to prolonged simulated microgravity and subsequent recovery would lead to increased oxidative damage and altered expression of genes involved in the oxidative response. To test this hypothesis, we examined hearts of male (three and nine months of age) and female (3 months of age) Long–Evans rats that underwent HU for various durations up to 90 days and reambulated up to 90 days post-HU. Results indicate sex-dependent changes in oxidative damage marker 8-hydroxydeoxyguanosine (8-OHdG) and antioxidant gene expression in left ventricular tissue. Three-month-old females displayed elevated 8-OHdG levels after 14 days of HU while age-matched males did not. In nine-month-old males, there were no differences in 8-OHdG levels between HU and normally loaded control males at any of the timepoints tested following HU. RNAseq analysis of left ventricular tissue from nine-month-old males after 14 days of HU revealed upregulation of pathways involved in pro-inflammatory signaling, immune cell activation and differential expression of genes associated with cardiovascular disease progression. Taken together, these findings provide a rationale for targeting antioxidant and immune pathways and that sex differences should be taken into account in the development of countermeasures to maintain cardiovascular health in space. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Body weights of animals included in this study. NL: normally loaded; HU: hindlimb unloading; Rel: reambulated (reloading). The HU group was compared to their age- and sex-matched NL controls by Student’s <span class="html-italic">t</span>-test. Sample sizes: N = 5–8/group for all groups except for 90D older males, where N = 3. Values depicted are means and standard deviation. * Significant at <span class="html-italic">p</span> &lt; 0.05 by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 2
<p>8-hydroxydeoxyguanosine (8-OHdG) levels in left ventricular tissue as measured by ELISA. NL: normally loaded control, HU: hindlimb unloading, 7D Rel: 90D HU + 7D reloading and normally loaded control (NL). The HU groups were compared to their age- and sex-matched NL controls by Student’s <span class="html-italic">t</span>-test. Sample sizes: N = 5–8/group for all groups except for 90D older males, where N = 3. Values depicted are means and standard deviation. * Significant at <span class="html-italic">p</span> &lt; 0.05 by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Transcript levels of (<b>A</b>) <span class="html-italic">Nfe2l2</span>, (<b>B</b>) <span class="html-italic">Sod1</span>, (<b>C</b>) <span class="html-italic">Sod2</span>, and (<b>D</b>) <span class="html-italic">Sirt1</span> in left ventricular wall as measured by qPCR. Values depicted are mean fold changes relative to young male control at 14 days of treatment as determined by the ΔΔCt method. Errors bars show upper and lower ranges. NL: normally loaded control, HU: hindlimb unloading, 90D Rel: 90D HU + 90D reloading and NL control. Sample sizes: N = 3–7/group. * Significant at <span class="html-italic">p</span> &lt; 0.05 by Student’s <span class="html-italic">t</span>-test by comparing HU with age- and sex-matched NL control.</p>
Full article ">Figure 4
<p>PCA plot of transcriptomic data from older males that underwent 14 days of HU (gray triangles) and corresponding NL controls (black circles). Animal ID is indicated by the letter “R” succeeded by numbers.</p>
Full article ">Figure 5
<p>Heatmap showing normalized counts of differentially expressed genes from older male 14D NL and HU groups. Each cell corresponds to a gene. Red: Upregulated in 14D HU relative to NL group. Blue: Downregulated in 14D HU relative to NL group. Magnitude of upregulation or downregulation is proportional to the intensity of red or blue. Deepest red: most upregulated; deepest blue: most downregulated.</p>
Full article ">Figure 6
<p>Top 10 upregulated and downregulated genes in older male 14D HU relative to NL groups. Log2 FC: Log2 fold change. Refer to <a href="#genes-15-00975-t001" class="html-table">Table 1</a> for full list of DEGs.</p>
Full article ">Figure 7
<p>Select enriched gene ontology (GO) terms for biological processes in older male 14D HU group relative to NL group. Gene count refers to the number of DEGs that matched the GO term. The vertical bar represents the color scale of the FDR with black representing the lowest FDR. Refer to <a href="#app1-genes-15-00975" class="html-app">Table S2</a> for the full list of GO terms.</p>
Full article ">
20 pages, 4383 KiB  
Article
Methanolic Extract of Phoenix Dactylifera Confers Protection against Experimental Diabetic Cardiomyopathy through Modulation of Glucolipid Metabolism and Cardiac Remodeling
by Laaraib Nawaz, David J. Grieve, Humaira Muzaffar, Arslan Iftikhar and Haseeb Anwar
Cells 2024, 13(14), 1196; https://doi.org/10.3390/cells13141196 - 15 Jul 2024
Viewed by 410
Abstract
The incidence of cardiovascular disorders is continuously rising, and there are no effective drugs to treat diabetes-associated heart failure. Thus, there is an urgent need to explore alternate approaches, including natural plant extracts, which have been successfully exploited for therapeutic purposes. The current [...] Read more.
The incidence of cardiovascular disorders is continuously rising, and there are no effective drugs to treat diabetes-associated heart failure. Thus, there is an urgent need to explore alternate approaches, including natural plant extracts, which have been successfully exploited for therapeutic purposes. The current study aimed to explore the cardioprotective potential of Phoenix dactylifera (PD) extract in experimental diabetic cardiomyopathy (DCM). Following in vitro phytochemical analyses, Wistar albino rats (N = 16, male; age 2–3 weeks) were fed with a high-fat or standard diet prior to injection of streptozotocin (35 mg/kg i.p.) after 2 months and separation into the following four treatment groups: healthy control, DCM control, DCM metformin (200 mg/kg/day, as the reference control), and DCM PD treatment (5 mg/kg/day). After 25 days, glucolipid and myocardial blood and serum markers were assessed along with histopathology and gene expression of both heart and pancreatic tissues. The PD treatment improved glucolipid balance (FBG 110 ± 5.5 mg/dL; insulin 17 ± 3.4 ng/mL; total cholesterol 75 ± 8.5 mg/dL) and oxidative stress (TOS 50 ± 7.8 H2O2equiv./L) in the DCM rats, which was associated with preserved structural integrity of both the pancreas and heart compared to the DCM control (FBG 301 ± 10 mg/dL; insulin 27 ± 3.4 ng/mL; total cholesterol 126 ± 10 mg/dL; TOS 165 ± 12 H2O2equiv./L). Gene expression analyses revealed that PD treatment upregulated the expression of insulin signaling genes in pancreatic tissue (INS-I 1.69 ± 0.02; INS-II 1.3 ± 0.02) and downregulated profibrotic gene expression in ventricular tissue (TGF-β 1.49 ± 0.04) compared to the DCM control (INS-I 0.6 ± 0.02; INS-II 0.49 ± 0.03; TGF-β 5.7 ± 0.34). Taken together, these data indicate that Phoenix dactylifera may offer cardioprotection in DCM by regulating glucolipid balance and metabolic signaling. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Consort diagram showing the protocol for the in vivo experimental trial.</p>
Full article ">Figure 2
<p><span class="html-italic">Phoenix dactylifera</span> metabolic extract showing an equivalent antihyperglycemic activity to acarbose, which is mediated by p-coumaric acid as its active bio-constituent. α-Amylase inhibition assay with comparison between PD and acarbose (Acr) as the standard drug control: (<b>A</b>) quantification of IC50; (<b>B</b>) concentration-dependent inhibition of α-amylase inhibition, mean ± SEM, N = 3; (<b>C</b>) representative HPLC trace showing detection of p-coumaric acid in PD extract signified by the indicated peak.</p>
Full article ">Figure 3
<p><span class="html-italic">Phoenix dactylifera</span> treatment restored glucolipid metabolic dysfunction in DCM: (<b>A</b>) progression of fasting blood glucose levels in healthy control and DCM control rats treated with or without PD or metformin. After 25 days, rats were sacrificed for serum analysis of (<b>B</b>) glucose, (<b>C</b>) HbA1c, (<b>D</b>) insulin, (<b>E</b>) total cholesterol, (<b>F</b>) triglyceride, (<b>G</b>) HDL-cholesterol, (<b>H</b>) LDL-cholesterol (<b>I</b>) cardiac risk ratio, and (<b>J</b>) cardioprotective index. Data are Mean ± SEM, N = 4. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p><span class="html-italic">Phoenix dactylifera</span> activates insulin signaling pathway genes and transcriptional factors and improves pancreatic islet morphology in DCM. Gene expression of (<b>A</b>) <span class="html-italic">INS-I</span>, (<b>B</b>) <span class="html-italic">INS-II</span>, (<b>C</b>) <span class="html-italic">PDX-1</span>, (<b>D</b>) <span class="html-italic">MAFA</span>, and (<b>E</b>) <span class="html-italic">GLUT-2</span> in the pancreas of the healthy control and DCM control rats treated with or without PD or metformin, by RT-qPCR, with normalization to β-actin as housekeeping control. Data are mean ± SEM, N = 4. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05. (<b>F</b>) Representative H&amp;E-stained histopathology sections (40× magnification) showing normal histological features islets of Langerhans, with scattered β cells (yellow arrows) and red blood cells (red arrows) visible in the vicinity (EX: exocrine pancreas) in healthy rat pancreases, which were disrupted in the DCM rat pancreases (deformed boundary and selective β-cell destruction) and restored by PD or metformin treatment. Scale bar = 20 µm.</p>
Full article ">Figure 5
<p><span class="html-italic">Phoenix dactylifera</span> treatment reduced myocardial oxidative stress in the DCM and protects against injury. The DCM rats were treated with or without PD or metformin for 25 days together with the healthy controls prior to sacrifice and collection of myocardial tissue for analysis of (<b>A</b>) pro-BNP, (<b>B</b>,<b>C</b>) ECG, (<b>D</b>–<b>F</b>) myocardial enzymes, (<b>G</b>) total antioxidant capacity, and (<b>H</b>,<b>I</b>) oxidative stress markers. Data are the means ± SEM, N = 4. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p><span class="html-italic">Phoenix dactylifera</span> treatment reduces proinflammatory and profibrotic gene expression and restores deranged myocardial structure in DCM. Gene expressions of (<b>A</b>) <span class="html-italic">NF-kB</span>, (<b>B</b>) <span class="html-italic">TGF-β</span>, and (<b>C</b>) <span class="html-italic">TNF-α</span> in the myocardium of healthy control and DCM control rats treated with or without PD or metformin, by RT-qPCR, with normalization to GAPDH as the housekeeping control. (<b>D</b>) Cardiomyocyte diameter measured with ImageJ software in a representative histological section of the ventricle (H&amp;E). (<b>E</b>) Relative organ weight of heart (mg/g). Data are the means ± SEM, N = 4. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.05. (<b>F</b>) Representative H&amp;E-stained histopathology sections (40× magnification) showing normal histological features in healthy rat myocardiums characterized by centrally located nuclei in striated fibers (yellow arrows) and uniform intercalated discs (green arrows), which were disrupted in the DCM rat myocardium (focal degeneration, white arrows; edema, blue stars) together with evidence of fibroblasts (blue arrows), fibrosis (white stars), lymphocyte infiltration (red arrows), hemorrhage (black arrows), and cardiomyocyte diameter (yellow straight lines), restored to different degrees by PD or metformin treatment. Scale bar = 20 µm.</p>
Full article ">Figure 7
<p>PCA inhibits in vitro myofibroblast differentiation. (<b>A</b>) HCF were treated with a range of concentrations of PCA for 3 h prior to analysis of cytotoxicity by MTT assay. HCFs were then stimulated with hyperglycemia and TGF-β with or without PCA compared to vehicle control for 48 h followed by quantifications of CTGF and α-SMA (<b>B</b>,<b>C</b>) mRNA expressions and (<b>D</b>–<b>F</b>) protein expression, by qRT-PCR and Western blot, respectively, with normalization to β-actin as the housekeeping control. Data are Mean ± SEM, N = 3 **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 8
<p>Summary of the overall effects of PD extract on diabetic cardiomyopathy.</p>
Full article ">
18 pages, 4617 KiB  
Article
Luteolin-7-O-β-d-Glucuronide Attenuated Cerebral Ischemia/Reperfusion Injury: Involvement of the Blood–Brain Barrier
by Xing Fan, Jintao Song, Shuting Zhang, Lihui Lu, Fang Lin, Yu Chen, Shichang Li, Xinxin Jin and Fang Wang
Biomedicines 2024, 12(6), 1366; https://doi.org/10.3390/biomedicines12061366 - 19 Jun 2024
Viewed by 603
Abstract
Ischemic stroke is a common cerebrovascular disease with high mortality, high morbidity, and high disability. Cerebral ischemia/reperfusion injury seriously affects the quality of life of patients. Luteolin-7-O-β-d-glucuronide (LGU) is a major active flavonoid compound extracted from Ixeris sonchifolia (Bge.) Hance, a Chinese medicinal [...] Read more.
Ischemic stroke is a common cerebrovascular disease with high mortality, high morbidity, and high disability. Cerebral ischemia/reperfusion injury seriously affects the quality of life of patients. Luteolin-7-O-β-d-glucuronide (LGU) is a major active flavonoid compound extracted from Ixeris sonchifolia (Bge.) Hance, a Chinese medicinal herb mainly used for the treatment of coronary heart disease, angina pectoris, cerebral infarction, etc. In the present study, the protective effect of LGU on cerebral ischemia/reperfusion injury was investigated in an oxygen–glucose deprivation/reoxygenation (OGD/R) neuronal model and a transient middle cerebral artery occlusion (tMCAO) rat model. In in vitro experiments, LGU was found to improve the OGD/R-induced decrease in neuronal viability effectively by the MTT assay. In in vivo experiments, neurological deficit scores, infarction volume rates, and brain water content rates were improved after a single intravenous administration of LGU. These findings suggest that LGU has significant protective effects on cerebral ischemia/reperfusion injury in vitro and in vivo. To further explore the potential mechanism of LGU on cerebral ischemia/reperfusion injury, we performed a series of tests. The results showed that a single administration of LGU decreased the content of EB and S100B and ameliorated the abnormal expression of tight junction proteins ZO-1 and occludin and metalloproteinase MMP-9 in the ischemic cerebral cortex of the tMCAO 24-h injury model. In addition, LGU also improved the tight junction structure between endothelial cells and the degree of basement membrane degradation and reduced the content of TNF-α and IL-1β in the brain tissue. Thereby, LGU attenuated cerebral ischemia/reperfusion injury by improving the permeability of the blood–brain barrier. The present study provides new insights into the therapeutic potential of LGU in cerebral ischemia. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Establishment of the OGD/R model in rat cerebral cortical neurons. (<b>A</b>): OGD/R:12 h/36 and 48 h. (<b>B</b>): OGD/R: 16 h/48 and 72 h. Data are expressed as mean ± S.E.M. (n = 3). ** <span class="html-italic">p</span> &lt; 0.01 *** <span class="html-italic">p</span> &lt; 0.000 vs. the ctrl group.</p>
Full article ">Figure 2
<p>Protective effect of LGU on the OGD/R-induced decrease in cell viability in rat primary cortical neurons. (<b>A</b>): OGD/R:12 h/36 h. (<b>B</b>): OGD/R: 12 h/48 h. (<b>C</b>): OGD/R:16 h/48 h. (<b>D</b>): OGD/R: 16 h/72 h.Data are expressed as mean ± S.E.M. (n = 3). ### <span class="html-italic">p</span> &lt; 0.001 vs. the ctrl group, * <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; 0.000 vs. OGD/R group.</p>
Full article ">Figure 3
<p>Establishment of the tMCAO rat model. (<b>A</b>): Neurological function score. (<b>B</b>): Cerebral infarct volume. (<b>C</b>): Brain water content. (<b>D</b>): Representative cerebral infarction image. Data are expressed as the mean ± S.E.M. n = 9–10. ### <span class="html-italic">p</span> &lt; 0.001, compared with the sham group; ★ <span class="html-italic">p</span> &lt; 0.05, ★★★ <span class="html-italic">p</span> &lt; 0.001 compared with the tMCAO 12 h group.</p>
Full article ">Figure 4
<p>Protective effect of LGU on ischemic brain injury in tMCAO rats. (<b>A</b>): Neurological function score. (<b>B</b>): Cerebral infarct volume. (<b>C</b>): Brain water content. (<b>D</b>): Representative cerebral infarction image. Data are expressed as the mean ± S.E.M. n = 7–9. ### <span class="html-italic">p</span> &lt; 0.001, compared with the sham group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared with the tMCAO model group.</p>
Full article ">Figure 5
<p>Effect of LGU on the permeability of Evans blue in tMCAO rats. (<b>A</b>): Representative images of EB leakage in the cerebral cortex. (<b>B</b>): Quantitative analysis of EB leakage. Data are expressed as the mean ± S.E.M.; n = 6–7. ### <span class="html-italic">p</span> &lt; 0.001, compared with the sham group; * <span class="html-italic">p</span> &lt; 0.05, compared with the model group.</p>
Full article ">Figure 6
<p>Effect of LGU on the content of serum S100B in tMCAO rats. Data are expressed as mean ± S.E.M. n = 7. # <span class="html-italic">p</span> &lt; 0.05, compared with the sham group; * <span class="html-italic">p</span> &lt; 0.05, compared with the model group.</p>
Full article ">Figure 7
<p>Effects of LGU on the ultrastructural changes in neurons in tMCAO rats: (<b>A</b>–<b>E</b>): 20,000× and (<b>A1</b>–<b>E1</b>): 40,000×. Mitochondria (red arrowhead) were observed. (<b>A</b>,<b>A1</b>): Sham; (<b>B</b>,<b>B1</b>): model; (<b>C</b>,<b>C1</b>): LGU (0.24 mg/kg); (<b>D</b>,<b>D1</b>): LGU (0.72 mg/kg); and (<b>E</b>,<b>E1</b>): LGU (2.16 mg/kg).</p>
Full article ">Figure 8
<p>Effects of LGU on the ultrastructural changes in the blood–brain barrier in tMCAO rats. The photomicrographs in (<b>A1</b>), (<b>B1</b>), (<b>C1</b>), and (<b>D1</b>) exhibit higher magnification images of the areas shown in frames in (<b>A</b>), (<b>B</b>), (<b>C</b>), and (<b>D</b>), respectively. The basement membrane (blue arrowhead) and tight junction (red arrowhead) were observed. (<b>A</b>,<b>A1</b>): Sham; (<b>B</b>,<b>B1</b>): model; (<b>C</b>,<b>C1</b>): LGU (0.24 mg/kg); (<b>D</b>,<b>D1</b>): LGU (0.72 mg/kg); (<b>E</b>,<b>E1</b>): LGU (2.16 mg/kg); (<b>A</b>–<b>E</b>): 20,000×; and (<b>A1</b>–<b>E1</b>): 40,000×.</p>
Full article ">Figure 9
<p>Effects of LGU on the expression of tight junction-related proteins in the ischemic cerebral cortex. (<b>A</b>): The expression of ZO-1 and Occludin of tignt junction proteins was examined by western blot analysis. (<b>B</b>): The relative expression of ZO-1 was normalized to β-actin. (<b>C</b>): The relative expression of Occludin was normalized to β-actin. Total protein was extracted from the ischemic cortex at 22 h of reperfusion after 2 h MCAO. n = 3. ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, compared with the sham group; ** <span class="html-italic">p</span> &lt; 0.01, compared with the model group.</p>
Full article ">Figure 10
<p>Effects of LGU on the expression of matrix metalloproteinase in the ischemic cerebral cortex. Expression levels of target proteins were detected by western blotting. (<b>A</b>): The expression of MMP-9 was examined by western blot analysis. (<b>B</b>): The expression of MMP-2 was examined by western blot analysis. Total protein was extracted from the ischemic cortex at 22 h of reperfusion after 2 h of MCAO. n = 3. ### <span class="html-italic">p</span> &lt; 0.001, compared with the sham group; * <span class="html-italic">p</span> &lt; 0.05, compared with the model group.</p>
Full article ">Figure 11
<p>Effects of LGU on the content of TNF-α and IL-1β in the ischemic cerebral cortex of rats. (<b>A</b>): Effects of LGU on the content of TNF-α in the ischemic cerebral cortex of rats. (<b>B</b>): Effects of LGU on the content of IL-1β in the ischemic cerebral cortex of rats. The effects of LGU on tissue homogenate expression of TNF-α and IL-1β (pg/mg) at 22 h of reperfusion after 2 h of MCAO. Data are expressed as the mean ± S.E.M. n = 7. ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, compared with the sham group; <span class="html-italic">* p</span> &lt; 0.05, compared with the model group.</p>
Full article ">
23 pages, 5752 KiB  
Article
A Double-Humanized Mouse Model for Studying Host Gut Microbiome–Immune Interactions in Gulf War Illness
by Dipro Bose, Punnag Saha, Subhajit Roy, Ayushi Trivedi, Madhura More, Nancy Klimas, Ashok Tuteja and Saurabh Chatterjee
Int. J. Mol. Sci. 2024, 25(11), 6093; https://doi.org/10.3390/ijms25116093 - 31 May 2024
Viewed by 554
Abstract
Unraveling the multisymptomatic Gulf War Illness (GWI) pathology and finding an effective cure have eluded researchers for decades. The chronic symptom persistence and limitations for studying the etiologies in mouse models that differ significantly from those in humans pose challenges for drug discovery [...] Read more.
Unraveling the multisymptomatic Gulf War Illness (GWI) pathology and finding an effective cure have eluded researchers for decades. The chronic symptom persistence and limitations for studying the etiologies in mouse models that differ significantly from those in humans pose challenges for drug discovery and finding effective therapeutic regimens. The GWI exposome differs significantly in the study cohorts, and the above makes it difficult to recreate a model closely resembling the GWI symptom pathology. We have used a double engraftment strategy for reconstituting a human immune system coupled with human microbiome transfer to create a humanized-mouse model for GWI. Using whole-genome shotgun sequencing and blood immune cytokine enzyme linked immunosorbent assay (ELISA), we show that our double humanized mice treated with Gulf War (GW) chemicals show significantly altered gut microbiomes, similar to those reported in a Veteran cohort of GWI. The results also showed similar cytokine profiles, such as increased levels of IL-1β, IL-6, and TNF R-1, in the double humanized model, as found previously in a human cohort. Further, a novel GWI Veteran fecal microbiota transfer was used to create a second alternative model that closely resembled the microbiome and immune-system-associated pathology of a GWI Veteran. A GWI Veteran microbiota transplant in humanized mice showed a human microbiome reconstitution and a systemic inflammatory pathology, as reflected by increases in interleukins 1β, 6, 8 (IL-1β, IL-6, IL-8), tumor necrosis factor receptor 1 (TNF R-1), and endotoxemia. In conclusion, though preliminary, we report a novel in vivo model with a human microbiome reconstitution and an engrafted human immune phenotype that may help to better understand gut–immune interactions in GWI. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Establishment of human gut bacteria in NSG-CD34+ mice. (<b>A</b>) Box plots showing α-diversities (Chao 1) of gut bacteriome in NSG_Control group (mice administered with the vehicle), NSG_ABX Treatment (mice administered with antibiotic cocktail via oral gavage for 12 days), and NSG_Hu-FMT (mice administered with human fecal microbiota transfer after gut bacteriome depletion with antibiotic cocktail). (<b>B</b>) β-Diversity analysis (Bray–Curtis) of NSG_Control, NSG_ABX Treatment, and NSG_Hu-FMT groups. (<b>C</b>) Stacked bar representation of relative abundance of gut bacteriome at the phylum level in NSG_Control, NSG_ABX Treatment, and NSG_Hu-FMT groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 2
<p>Bar graph representation of relative abundances of gut bacteriome at the genus level in NSG_Control, NSG_ABX Treatment, and NSG_Hu-FMT groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 3
<p>Altered expressions of systemic proinflammation biomarkers during establishment of human gut bacteria in NSG-CD34+ mice. Bar graph representation of systemic cytokines (<b>A</b>) IL-1β, (<b>B</b>) IL-6, (<b>C</b>) IL-8, and (<b>D</b>) TNF R-1 in NSG_Control, NSG_ABX Treatment, and NSG_Hu-FMT groups. (<b>E</b>) Bar graph representation of serum endotoxemia, measured by LAL Assay in NSG_Control, NSG_ABX Treatment, and NSG_Hu-FMT groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant. NS denotes non-significant change.</p>
Full article ">Figure 4
<p>Exposure to representative GW chemicals altered gut bacteriome profile in NSG-CD34+ mice with established human gut bacteria. (<b>A</b>) Box plots showing α-diversities of gut bacteriome (Chao 1) in NSG_Hu-FMT (mice administered with human fecal microbiota transfer after gut bacteriome depletion with antibiotic cocktail) and NSG_Hu-FMT+GWI (mice administered with representative GW chemicals pyridostigmine bromide and permethrin for 15 days after gut bacteriome depletion with antibiotic cocktail and human fecal microbiota transfer). (<b>B</b>) β-Diversity analysis (Bray–Curtis) of NSG_Hu-FMT and NSG_Hu-FMT+GWI groups. (<b>C</b>) Stacked bar representation of relative abundance of gut bacteriome at the phylum level in NSG_Hu-FMT and NSG_Hu-FMT+GWI groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 5
<p>Bar graph representation of relative abundances of gut bacteriome at the genus level in NSG_Hu-FMT and NSG_Hu-FMT+GWI groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 6
<p>Exposure to representative GW chemicals altered gut resistome profile in NSG-CD34+ mice with established human gut bacteria. (<b>A</b>) Box plots showing α-diversities (Chao 1) of gut resistome in NSG_Hu-FMT (mice administered with human fecal microbiota transfer after gut bacteriome depletion with antibiotic cocktail) and NSG_Hu-FMT+GWI (mice administered with representative GW chemicals pyridostigmine bromide and permethrin for 15 days after gut bacteriome depletion with antibiotic cocktail and human fecal microbiota transfer). (<b>B</b>) β-Diversity analysis (Bray–Curtis) of NSG_Hu-FMT and NSG_Hu-FMT+GWI groups. (<b>C</b>) Bar graph representation of relative abundances of altered antibiotic resistance genes in NSG_Hu-FMT and NSG_Hu-FMT+GWI groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 7
<p>Altered expression of systemic proinflammation biomarkers after administration with representative GW chemicals in NSG-CD34+ mice with established human gut bacteria. Bar graph representation of systemic cytokines (<b>A</b>) IL-1β, (<b>B</b>) IL-6, (<b>C</b>) IL-8, and (<b>D</b>) TNF R-1 in NSG_Hu-FMT and NSG_Hu-FMT+GWI groups. (<b>E</b>) Bar graph representation of serum endotoxemia, measured by LAL assay in NSG_Hu-FMT and NSG_Hu-FMT+GWI groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 8
<p>Administration with human fecal microbiota transfer from GWI Veteran’s stool sample altered gut bacteriome profile in NSG-CD34+ mice. (<b>A</b>) Box plots showing α-diversities of gut bacteriome (Chao 1) in NSG_Hu-FMT (mice administered with human fecal microbiota transfer after gut bacteriome depletion with antibiotic cocktail) and NSG_GWIV (mice administered with human fecal microbiota transfer from GWI Veteran’s stool sample after gut bacteriome depletion with antibiotic cocktail). (<b>B</b>) β-Diversity analysis (Bray–Curtis) of NSG_Hu-FMT and NSG_GWIV groups. (<b>C</b>) Stacked bar representation of relative abundances of gut bacteriome at the phylum level in NSG_Hu-FMT and NSG_GWIV groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 9
<p>Bar graph representation of relative abundances of gut bacteriome at the genus level in NSG_Hu-FMT and NSG_GWIV groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 10
<p>Administration with human fecal microbiota transfer from GWI Veteran’s stool sample altered gut resistome profile in NSG-CD34+ mice. (<b>A</b>) Box plots showing α-diversities (Chao 1) of gut resistome in NSG_Hu-FMT (mice administered with human fecal microbiota transfer after gut bacteriome depletion with antibiotic cocktail) and NSG_GWIV (mice administered with human fecal microbiota transfer from GWI Veteran’s stool sample after gut bacteriome depletion with antibiotic cocktail. (<b>B</b>) β-Diversity analysis (Bray–Curtis) of NSG_Hu-FMT and NSG_GWIV groups. (<b>C</b>) Bar graph representation of relative abundances of altered antibiotic resistance genes in NSG_Hu-FMT and NSG_GWIV groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">Figure 11
<p>Altered expressions of systemic proinflammation biomarkers, after human fecal microbiota transfer from GWI Veteran’s stool sample, in NSG-CD34+ mice. Bar graph representation of systemic cytokines (<b>A</b>) IL-1β, (<b>B</b>) IL-6, (<b>C</b>) IL-8, and (<b>D</b>) TNF R-1 in NSG_Hu-FMT and NSG_GWIV groups. (<b>E</b>) Bar graph representation of serum endotoxemia, measured by LAL assay in NSG_Hu-FMT and NSG_GWIV groups; <span class="html-italic">p</span> &lt; 0.05 was considered as statistically significant.</p>
Full article ">
15 pages, 3049 KiB  
Article
Mitochondrial Aldehyde Dehydrogenase 2 (ALDH2) Protects against Binge Alcohol-Mediated Gut and Brain Injury
by Bipul Ray, Wiramon Rungratanawanich, Karli R. LeFort, Saravana Babu Chidambaram and Byoung-Joon Song
Cells 2024, 13(11), 927; https://doi.org/10.3390/cells13110927 - 28 May 2024
Viewed by 648
Abstract
Mitochondrial aldehyde dehydrogenase-2 (ALDH2) metabolizes acetaldehyde to acetate. People with ALDH2 deficiency and Aldh2-knockout (KO) mice are more susceptible to alcohol-induced tissue damage. However, the underlying mechanisms behind ALDH2-related gut-associated brain damage remain unclear. Age-matched young female Aldh2-KO and C57BL/6J wild-type [...] Read more.
Mitochondrial aldehyde dehydrogenase-2 (ALDH2) metabolizes acetaldehyde to acetate. People with ALDH2 deficiency and Aldh2-knockout (KO) mice are more susceptible to alcohol-induced tissue damage. However, the underlying mechanisms behind ALDH2-related gut-associated brain damage remain unclear. Age-matched young female Aldh2-KO and C57BL/6J wild-type (WT) mice were gavaged with binge alcohol (4 g/kg/dose, three doses) or dextrose (control) at 12 h intervals. Tissues and sera were collected 1 h after the last ethanol dose and evaluated by histological and biochemical analyses of the gut and hippocampus and their extracts. For the mechanistic study, mouse neuroblast Neuro2A cells were exposed to ethanol with or without an Aldh2 inhibitor (Daidzin). Binge alcohol decreased intestinal tight/adherens junction proteins but increased oxidative stress-mediated post-translational modifications (PTMs) and enterocyte apoptosis, leading to elevated gut leakiness and endotoxemia in Aldh2-KO mice compared to corresponding WT mice. Alcohol-exposed Aldh2-KO mice also showed higher levels of hippocampal brain injury, oxidative stress-related PTMs, and neuronal apoptosis than the WT mice. Additionally, alcohol exposure reduced Neuro2A cell viability with elevated oxidative stress-related PTMs and apoptosis, all of which were exacerbated by Aldh2 inhibition. Our results show for the first time that ALDH2 plays a protective role in binge alcohol-induced brain injury partly through the gut–brain axis, suggesting that ALDH2 is a potential target for attenuating alcohol-induced tissue injury. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Binge alcohol exposure caused more intestinal disintegration and increased serum FITC-D4 and endotoxin (LPS) levels in <span class="html-italic">Aldh2</span>-KO mice compared to WT mice. (<b>A</b>) Representative H&amp;E-stained histology images showed more ruptured intestinal villi structures in alcohol-exposed <span class="html-italic">Aldh2</span>-KO mice than corresponding WT mice, (<b>B</b>) despite the similar levels of serum EtOH concentrations between both mouse strains 1 h after alcohol exposure. (<b>C</b>–<b>F</b>) Binge alcohol exposure significantly increased the levels of (<b>C</b>) serum FITC-D4, (<b>D</b>) endotoxin (LPS), (<b>E</b>) IL-6, and (<b>F</b>) TNF-α in <span class="html-italic">Aldh2</span>-KO mice compared to the WT counterparts (samples from n = 3~5/group). Data were analyzed by two-way ANOVA, where ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Binge alcohol significantly elevated intestinal apoptosis in <span class="html-italic">Aldh2</span>-KO mice compared to WT mice. (<b>A</b>) Representative images of TUNEL staining where the red arrows indicate TUNEL-positive cells, (<b>B</b>) quantification of TUNEL-positive cells, and (<b>C</b>) Caspase-3 activity assay results of the enterocyte extracts from the designated groups, revealing significantly higher levels of enterocyte apoptosis in alcohol-exposed <span class="html-italic">Aldh2</span>-KO mice than in the WT counterparts (n = 3~5/group). Data were analyzed by two-way ANOVA, where * <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.</p>
Full article ">Figure 3
<p>Binge alcohol exposure elevated intestinal apoptosis, oxidative stress-related PTMs, and degradation of gut TJ/AJ proteins in <span class="html-italic">Aldh2</span>-KO mice. (<b>A</b>) Representative immunoblot images of oxidative stress, PTMs, and apoptosis marker proteins in the gut extracts, as indicated. (<b>B</b>) Representative immunoblot images of intestinal TJ/AJ proteins, E-Cadherin, Claudin-1, β-Catenin, Occludin, and α-Tubulin, along with Aldh2 and Gapdh, for the indicated mouse groups (extracts from n = 3~5/group). (<b>C</b>) Densitometric quantitation of each protein level compared to Gapdh loading controls. The data were analyzed by two-way ANOVA, where * <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, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Binge alcohol increased brain damage in <span class="html-italic">Aldh2</span>-KO mice compared to WT mice. (<b>A</b>) The damaged neuronal cells detected by F.-J.C. staining were shown in bright green, as some were marked with red arrows in the hippocampus dentate gyrus region of mouse brains (n = 3/group, two separate preparations). DAPI was used to stain all neuronal cells. (<b>B</b>) Fluorescence intensity quantification. Data were analyzed by two-way ANOVA, where **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Binge alcohol exposure significantly increased the levels of acrolein-protein adducts and cleaved caspase-3, along with Caspase-3 activity, in the hippocampus extracts of alcohol-exposed <span class="html-italic">Aldh2</span>-KO mice compared to the WT counterparts (extracts from n = 3~5/group). (<b>A</b>) Representative immunoblot images and (<b>B</b>) densitometric quantitation of the indicated hippocampal protein, compared to Gapdh loading controls, and caspase-3 activity. These results were analyzed by two-way ANOVA, where * <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.</p>
Full article ">Figure 6
<p>ALDH2 suppression enhanced oxidative PTMs and apoptosis of Neuro2A cells. (<b>A</b>) The rates of cell viability in Neuro2A cells exposed to an ALDH2 inhibitor (Daidzin) at 20 or 40 µM and 12.5 or 25 mM EtOH. (<b>B</b>) Representatives immunoblot images of ALDH2 and oxidative PTMs. (<b>C</b>) ALDH2 activity and densitometric quantitation of the indicated protein relative to Gapdh loading controls (extracts from n = 3–5/group, repeated twice). Data were analyzed by one-way ANOVA, where * <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.</p>
Full article ">
11 pages, 1895 KiB  
Communication
Decreased Memory and Learning Ability Mediated by Bmal1/M1 Macrophages/Angptl2/Inflammatory Cytokine Pathway in Mice Exposed to Long-Term Blue Light Irradiation
by Keiichi Hiramoto, Sayaka Kubo, Keiko Tsuji, Daijiro Sugiyama and Hideo Hamano
Curr. Issues Mol. Biol. 2024, 46(5), 4924-4934; https://doi.org/10.3390/cimb46050295 - 18 May 2024
Viewed by 571
Abstract
Humans are persistently exposed to massive amounts of blue light via sunlight, computers, smartphones, and similar devices. Although the positive and negative effects of blue light on living organisms have been reported, its impact on learning and memory remains unknown. Herein, we examined [...] Read more.
Humans are persistently exposed to massive amounts of blue light via sunlight, computers, smartphones, and similar devices. Although the positive and negative effects of blue light on living organisms have been reported, its impact on learning and memory remains unknown. Herein, we examined the effects of widespread blue light exposure on the learning and memory abilities of blue light-exposed mice. Ten-week-old male ICR mice were divided into five groups (five mice/group) and irradiated with blue light from a light-emitting diode daily for 6 months. After 6 months of blue light irradiation, mice exhibited a decline in memory and learning abilities, assessed using the Morris water maze and step-through passive avoidance paradigms. Blue light-irradiated mice exhibited a decreased expression of the clock gene brain and muscle arnt-like 1 (Bmal1). The number of microglia and levels of M1 macrophage CC-chemokine receptor 7 and inducible nitric oxide synthase were increased, accompanied by a decrease in M2 macrophage arginase-1 levels. Levels of angiopoietin-like protein 2 and inflammatory cytokines interleukin-6, tumor necrosis factor-α, and interleukin-1β were elevated. Our findings suggest that long-term blue light exposure could reduce Bmal1 expression, activate the M1 macrophage/Angptl2/inflammatory cytokine pathway, induce neurodegeneration, and lead to a decline in memory. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of the experimental method.</p>
Full article ">Figure 2
<p>Effect of long-term blue light irradiation on body weight (<b>A</b>), motor activity (<b>B</b>) memory and learning ability (<b>C</b>,<b>D</b>) of mice. The Morris water maze test (<b>C</b>) and step-through passive avoidance test (<b>D</b>) were used to the assess the memory and learning ability of mice. Data values are expressed as the mean ± standard deviation (SD) derived from five specimens. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Effect of long-term blue light irradiation on the expression of Bmal1 (<b>A</b>), Cry1. (<b>B</b>), and Cry2 (<b>C</b>) in the hippocampus of mice. Western blot diagram of Bmal1, Cry1, and Cry2 with molecular weight markers (<b>D</b>). Data values are expressed as the mean ± standard deviation (SD) derived from five specimens. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Effect of long-term blue light irradiation on the expression of Iba1 (<b>A</b>), CCR7 (<b>B</b>), iNOS (<b>D</b>), and Arg-1 (<b>E</b>) in the hippocampus of mice. Western blot diagram of Iba1 and CCR7 with molecular weight markers (<b>C</b>). Data values are expressed as mean ± standard deviation (SD) derived from five specimens. <span class="html-italic">* p</span> &lt; 0.05; <span class="html-italic">*</span>* <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Effect of long-term blue light irradiation on the expression of Angptl2 (<b>A</b>), IL-6 (<b>B</b>), TNF-α (<b>C</b>), and IL-1β (<b>D</b>) in the hippocampus of the mice specimens. The values are expressed as means ± SD derived from five specimens. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Mechanism of the effect of long-term blue light irradiation on memory and learning ability in mice.</p>
Full article ">
16 pages, 3262 KiB  
Article
Restraint Stress-Induced Neutrophil Inflammation Contributes to Concurrent Gastrointestinal Injury in Mice
by Rina Munalisa, Te-Sheng Lien, Ping-Yeh Tsai, Der-Shan Sun, Ching-Feng Cheng, Wen-Sheng Wu, Chi-Cheng Li, Chi-Tan Hu, Kuo-Wang Tsai, Yungling Leo Lee, Yu-Chi Chou and Hsin-Hou Chang
Int. J. Mol. Sci. 2024, 25(10), 5261; https://doi.org/10.3390/ijms25105261 - 11 May 2024
Viewed by 1039
Abstract
Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we [...] Read more.
Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we employed the restraint stress mouse model, a well-established method to study the pathophysiological changes associated with psychological stress in mice. By orally administering gut-nonabsorbable Evans blue dye and monitoring its plasma levels, we were able to track the progression of gastrointestinal injury in live mice. Additionally, flow cytometry was utilized to assess the viability, death, and inflammatory status of splenic leukocytes, providing insights into the stress-induced impact on the innate immune system associated with stress-induced gastrointestinal injury. Our findings reveal that neutrophils represent the primary innate immune leukocyte lineage responsible for stress-induced inflammation. Splenic neutrophils exhibited elevated expression levels of the pro-inflammatory cytokine IL-1, cellular reactive oxygen species, mitochondrial burden, and cell death following stress challenge compared to other innate immune cells such as macrophages, monocytes, and dendritic cells. Regulated cell death analysis indicated that NETosis is the predominant stress-induced cell death response among other analyzed regulated cell death pathways. NETosis culminates in the formation and release of neutrophil extracellular traps, which play a crucial role in modulating inflammation by binding to pathogens. Treatment with the NETosis inhibitor GSK484 rescued stress-induced neutrophil extracellular trap release and gastrointestinal injury, highlighting the involvement of neutrophil extracellular traps in stress-induced gastrointestinal inflammation. Our results suggest that neutrophil NETosis could serve as a promising drug target for managing psychological stress-induced gastrointestinal injuries. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Restraint stress-induced gastrointestinal (GI) leakage is associated with the infiltration of innate immune leukocytes in the spleen of mice. (<b>A</b>) Experiment outline. (<b>B</b>) Plasma Evans blue levels, indicating the stress-induced GI leakage and injury levels. ND: not detected. (<b>C</b>) An example of the flow cytometry gating of Ly6G<sup>+</sup> leukocytes after 9 h stress; the same method is applied to the analyses in (<b>D</b>). In (<b>C</b>), the letters A and C represent the gated total live splenic leukocytes and the LY6G<sup>+</sup> cell population, respectively. (<b>D</b>) Relative cell-number levels of different leukocyte lineages (Ly6G<sup>+</sup> neutrophil, F4/80<sup>+</sup> macrophage, CD11b<sup>+</sup> monocyte, CD11c<sup>+</sup> dendritic cell) without (no stress groups) and with (stress groups) stress in the spleen of mice. The levels of respective leukocyte lineages in the no stress groups were normalized to one-fold. (<b>B</b>,<b>C</b>) * <span class="html-italic">p</span> &gt; 0.05, ** <span class="html-italic">p</span> &gt; 0.01, vs. no stress groups. <span class="html-italic">n</span> = 6 (3 experiments with a total of 6 mice per group). The mouse graph was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 1 January 2024.</p>
Full article ">Figure 2
<p>Restraint stress on the regulation of the cytokine expression of splenic innate immune leukocytes in mice. (<b>A</b>) Experiment outline. Blue: no stress groups; Red: stressed groups. (<b>B</b>) Relative levels of spleen splenocytes expressing pro-inflammatory cytokines IL-1β, TNF-α, IL-6, and anti-inflammatory cytokine IL-10, in mice with (stress groups) and without (no stress groups) 9 h stress. The levels of respective leukocyte lineages in the no stress groups were normalized to one-fold. (<b>C</b>) An example of flow cytometry gating for analyzing IL-1 expression in Ly6G<sup>+</sup> leukocytes; the same method is applied to the analyses in (<b>C</b>–<b>F</b>). (<b>C</b>–<b>F</b>) Relative expression levels of IL-1β (<b>D</b>), TNF-α (<b>E</b>), IL-6 (<b>F</b>), IL-10 (<b>G</b>) in different leukocyte lineages (Ly6G<sup>+</sup> neutrophil, F4/80<sup>+</sup> macrophage, CD11b<sup>+</sup> monocyte, CD11c<sup>+</sup> dendritic cell) without (no stress groups) and with (stress groups) stress. The expression percentage of tested cytokines was normalized to a baseline value of 1 for the respective leukocyte lineage population in the no stress groups. (<b>B</b>,<b>D</b>–<b>G</b>) * <span class="html-italic">p</span> &gt; 0.05, ** <span class="html-italic">p</span> &gt; 0.01, vs. no stress groups; <span class="html-italic">n</span> = 6 (3 experiments with a total of 6 mice per group). The mouse graph was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 1 January 2024.</p>
Full article ">Figure 3
<p>Relative levels of cell death, cellular ROS, and mitochondrial superoxide of splenic innate immune leukocytes. (<b>A</b>–<b>D</b>) Relative percentage of live and death cells of innate immune splenocytes, including (<b>A</b>) Ly6G<sup>+</sup> neutrophil, (<b>B</b>) F4/80<sup>+</sup> macrophage, (<b>C</b>) CD11b<sup>+</sup> monocyte, (<b>D</b>) CD11c<sup>+</sup> dendritic cell, in mice with (stress + groups) and without (stress − groups) stress. The levels of respective leukocyte lineages in the no stress groups were normalized to one-fold. (<b>E</b>–<b>G</b>) Relative levels of cellular ROS (<b>E</b>), and mitochondrial superoxide (<b>F</b>) without (no stress groups) and with (stress groups) stress in mice were analyzed. (<b>E</b>,<b>F</b>) The levels of respective leukocyte lineages in the no stress groups were normalized to one-fold. (<b>A</b>–<b>G</b>) * <span class="html-italic">p</span> &gt; 0.05, ** <span class="html-italic">p</span> &gt; 0.01, vs. respective no stress groups; <span class="html-italic">n</span> = 6 (3 experiments with a total of 6 mice per group). The mouse graph was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 1 January 2024.</p>
Full article ">Figure 4
<p>Restraint stress-induced cell death of splenic neutrophil in wild-type (<span class="html-italic">ATF3</span><sup>+/+</sup>) and ATF3 deficient (<span class="html-italic">ATF3<sup>−/−</sup></span>) mice. (<b>A</b>) Experiment outline. (<b>B</b>) Live and death cell percentage of splenic neutrophil from <span class="html-italic">ATF3</span><sup>+/+</sup> and <span class="html-italic">ATF3</span><sup>−/−</sup> mice without (stress − groups) and with (stress + groups) stress were analyzed by flow cytometry using Zombie NIR live/death analysis kit. Light blue represents the live cell population, while pink represents the dead cell population. (<b>C</b>–<b>H</b>) The regulated cell death (RCD) profiles, denoted by the alterations in the percentage of RCD-signal positive neutrophils in <span class="html-italic">ATF3</span><sup>+/+</sup> and <span class="html-italic">ATF3</span><sup>−/−</sup> mice under unstressed (stress − groups) and stressed (stress + groups) conditions, were assessed. The percentages of neutrophil RCD-marker<sup>+</sup> cells, including NETosis (<b>C</b>), apoptosis (<b>D</b>), autophagy (<b>E</b>), ferroptosis (<b>F</b>), necroptosis (<b>G</b>), and pyroptosis (<b>H</b>), were delineated. In each group, the signal from wild-type mice was normalized to one-fold; <span class="html-italic">n</span> = 6 (3 experiments with a total of 6 mice per group); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, significantly higher vs. without stress control groups; # <span class="html-italic">p</span> &lt; 0.05, significantly higher vs. wild-type (WT) control groups. The mouse graph was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 1 January 2024.</p>
Full article ">Figure 5
<p>Restraint stress-induced changes in splenic neutrophil in wild-type (<span class="html-italic">ATF3</span><sup>+/+</sup>) and ATF3 deficient (<span class="html-italic">ATF3<sup>−/−</sup></span>) mice. (<b>A</b>) Relative cell abundance, (<b>B</b>) mitochondrial mass, (<b>C</b>) mitochondrial membrane potential, and (<b>D</b>) cellular ROS levels in the spleen neutrophils of <span class="html-italic">ATF3</span><sup>+/+</sup> and <span class="html-italic">ATF3</span><sup>−/−</sup> mice, with (stress + groups) and without (stress − groups) stress, were analyzed by flow cytometry. Blue denotes the no-stress groups, while red indicates the stressed groups. <span class="html-italic">n</span> = 6 (3 experiments with a total of 6 mice per group); * <span class="html-italic">p</span> &lt; 0.05, vs. vehicle control (0 g/mL) groups; # <span class="html-italic">p</span> &lt; 0.05, significantly higher vs. wild-type (WT) control groups.</p>
Full article ">Figure 6
<p>Treatments of NETosis inhibitor GSK484 rescued restraint stress-induced GI leakage and pro-inflammatory changes in splenic neutrophil in mice. (<b>A</b>) Experiment outline. (<b>B</b>) Plasma Evans blue levels, (<b>C</b>) neutrophil cell death levels, (<b>D</b>) neutrophil NETosis levels, in the spleen of mice, with (stress + groups) or without (stress − groups) stress, and with (GSK484 + groups) or without (GSK484 − groups) GSK484 treatments, were analyzed; <span class="html-italic">n</span> = 6 (3 experiments with a total of 6 mice per group); # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, exacerbated vs. no stress control groups; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, rescued vs. vehicle control (0 g/mL, without GSK484 treatments) groups. (<b>B</b>) ND: not detected. (<b>C</b>) Light blue represents the live cell population, while pink represents the dead cell population. (<b>D</b>) The mouse graph was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>, accessed on 1 January 2024.</p>
Full article ">
11 pages, 2013 KiB  
Article
Optimizing Embryo Collection for Application of CRISPR/Cas9 System and Generation of Fukutin Knockout Rat Using This Method
by Dong-Won Seol, Byoung-Jin Park, Deog-Bon Koo, Ji-Su Kim, Yong-Hyun Jeon, Jae-Eon Lee, Joon-Suk Park, Hoon Jang and Gabbine Wee
Curr. Issues Mol. Biol. 2024, 46(5), 3752-3762; https://doi.org/10.3390/cimb46050234 - 23 Apr 2024
Viewed by 704
Abstract
Rat animal models are widely used owing to their relatively superior cognitive abilities and higher similarity compared with mouse models to human physiological characteristics. However, their use is limited because of difficulties in establishing embryonic stem cells and performing genetic modifications, and insufficient [...] Read more.
Rat animal models are widely used owing to their relatively superior cognitive abilities and higher similarity compared with mouse models to human physiological characteristics. However, their use is limited because of difficulties in establishing embryonic stem cells and performing genetic modifications, and insufficient embryological research. In this study, we established optimal superovulation and fertilized–egg transfer conditions, including optimal hormone injection concentration (≥150 IU/kg of PMSG and hCG) and culture medium (mR1ECM), to obtain high-quality zygotes and establish in vitro fertilization conditions for rats. Next, sgRNA with optimal targeting activity was selected by performing PCR analysis and the T7E1 assay, and the CRISPR/Cas9 system was used to construct a rat model for muscular dystrophy by inducing a deficiency in the fukutin gene without any off-target effect detected. The production of fukutin knockout rats was phenotypically confirmed by observing a drop-in body weight to one-third of that of the control group. In summary, we succeeded in constructing the first muscular dystrophy disease rat model using the CRISPR/CAS9 system for increasing future prospects of producing various animal disease models and encouraging disease research using rats. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Establishment of optimal superovulation of experimental embryonic system in rat animal model. (<b>A</b>) Comparison of superovulation rates according to doses of PMSG and hCG. (<b>B</b>) Comparison of dead zygotes according to doses of PMSG and hCG. *Asterisks indicate significant differences, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Construction of CRISPR/Cas9-based rat fukutin gene knockout system. (<b>A</b>) Target sequences recommended by CRISPR RGEN tools in rat fukutin gene exon. The target region is represented in red text, and the PAM sequence is represented in green text. sgRNA sequences are indicated in order as a, b, and c. (<b>B</b>) Verification of efficiency of designed sgRNA. The three recommended types of sgRNA and CRISPR/Cas9 vector were each transfected into rat fibroblasts, and 2 days later, gDNA was isolated and PCR and T7E1 analyses were performed. The arrow indicates that the gene targeting was successful.</p>
Full article ">Figure 3
<p>Production of fukutin gene-deficient rats and identification of target sequences. (<b>A</b>) T7E1 assay to identify fukutin-deficient rat litter. The arrow indicates that the gene targeting was successful. (<b>B</b>) Analysis of genetic defect types using DNA sequencing in the genomes of two founders 1 and 3. ∆ indicates deletion of DNA base and + indicates insertion of DNA base. Blue text indicates the target site, green text indicates the PAM sequence, and red text indicates the insertion of the DNA base.</p>
Full article ">Figure 4
<p>Comparison of morphology and skeletal muscle in founders. (<b>A</b>) The body weights of fukutin-deficient rats were compared at 3, 5, and 7 weeks, respectively. (<b>B</b>) Photograph of fukutin-deficient rats and WT at 3, 5 and 7 weeks. (<b>C</b>) Cross-sections of quadriceps muscles stained with H&amp;E. (<b>D</b>) Analysis of fiber diameters and size in founder 1 and WT. Data are the means ± SE and analyzed by sStudent’s <span class="html-italic">t</span>-test. Values with different superscripts denote a significant difference compared with other groups (* <span class="html-italic">p</span> &lt; 0.05). White arrows indicate fascicle diameter.</p>
Full article ">
21 pages, 4072 KiB  
Article
AAV-Mediated Restoration of Dystrophin-Dp71 in the Brain of Dp71-Null Mice: Molecular, Cellular and Behavioral Outcomes
by Ophélie Vacca, Faouzi Zarrouki, Charlotte Izabelle, Mehdi Belmaati Cherkaoui, Alvaro Rendon, Deniz Dalkara and Cyrille Vaillend
Cells 2024, 13(8), 718; https://doi.org/10.3390/cells13080718 - 20 Apr 2024
Viewed by 1421
Abstract
A deficiency in the shortest dystrophin-gene product, Dp71, is a pivotal aggravating factor for intellectual disabilities in Duchenne muscular dystrophy (DMD). Recent advances in preclinical research have achieved some success in compensating both muscle and brain dysfunctions associated with DMD, notably using exon [...] Read more.
A deficiency in the shortest dystrophin-gene product, Dp71, is a pivotal aggravating factor for intellectual disabilities in Duchenne muscular dystrophy (DMD). Recent advances in preclinical research have achieved some success in compensating both muscle and brain dysfunctions associated with DMD, notably using exon skipping strategies. However, this has not been studied for distal mutations in the DMD gene leading to Dp71 loss. In this study, we aimed to restore brain Dp71 expression in the Dp71-null transgenic mouse using an adeno-associated virus (AAV) administrated either by intracardiac injections at P4 (ICP4) or by bilateral intracerebroventricular (ICV) injections in adults. ICP4 delivery of the AAV9-Dp71 vector enabled the expression of 2 to 14% of brain Dp71, while ICV delivery enabled the overexpression of Dp71 in the hippocampus and cortex of adult mice, with anecdotal expression in the cerebellum. The restoration of Dp71 was mostly located in the glial endfeet that surround capillaries, and it was associated with partial localization of Dp71-associated proteins, α1-syntrophin and AQP4 water channels, suggesting proper restoration of a scaffold of proteins involved in blood–brain barrier function and water homeostasis. However, this did not result in significant improvements in behavioral disturbances displayed by Dp71-null mice. The potential and limitations of this AAV-mediated strategy are discussed. This proof-of-concept study identifies key molecular markers to estimate the efficiencies of Dp71 rescue strategies and opens new avenues for enhancing gene therapy targeting cognitive disorders associated with a subgroup of severely affected DMD patients. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Transduction territories of the AAV9-CAG-GFP vector following intracardiac administration: (<b>A</b>) GFP expression (green) in whole-brain sections of wild-type (left) and Dp71-null (right) mice after intracardiac injection at postnatal day 4 (ICP4) of AAV9-GFP vector (scale bar: 500 µm); (<b>B</b>,<b>C</b>) colocalization of GFP expression (green) and the neuronal marker NeuN (red) or astrocyte marker GFAP (red) in the hippocampus (<b>B</b>) and cerebellum (<b>C</b>). The nuclear marker, DAPI, is shown in blue (scale bar: 100 µm). Note that AAV9 intracardially injected at P4 widely transduced non-neuronal, mostly glial, cells in both wild-type and Dp71-null mice. SLM: <span class="html-italic">stratum lacunosum moleculare</span>; MCL: molecular cell layer.</p>
Full article ">Figure 2
<p>Dp71 re-expression following intracardiac injections at postnatal day 4 (ICP4). Mice were injected with the AAV9-CAG-GFP control vector (WT-C, Dp71-C) and AAV9-CBA-GFP-2A-Dp71 vector (Dp71-T). (<b>A</b>) Dp71 mRNA levels quantified by qPCR in the three groups of mice in the hippocampus, cortex and cerebellum, as indicated. (<b>B</b>) Dp71 protein levels quantified using Western blots in the three groups of mice in the hippocampus (HIP), cortex CX) and cerebellum (CBL). The image on the left shows an example of a Western blot with 4 mice from each group and the detection using the H4 antibody of dystrophins Dp427, Dp140 and Dp71 (green). Vinculin (red) was used as control protein for normalization. Importantly, note that all (n = 7) treated mice showed Dp71 protein rescue. (<b>C</b>) Dp71 localization by immunofluorescence staining using the pan-specific H4 antibody (red) on 12 µm cryosections. Images were taken at ×10 and ×0.5 magnifications (inserts: ×20 and ×1 magnifications) (scale bar: 100 µm). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span>&lt; 0.01, ns for non-significant, Mann–Whitney test.</p>
Full article ">Figure 3
<p>Expressions of AQP4 and α1-syntrophin along the walls of blood vessels after intracardiac administration (ICP4) of the AAV vectors. (<b>A</b>,<b>B</b>) For both antibodies, the immunofluorescent (IF) staining is shown in tissue sections of the hippocampus and cerebellum, as indicated, in Dp71-null mice injected with the AAV9-CBA-GFP-2A-Dp71 vector (Dp71-T) and with the control vector (Dp71-C, WT, C). (<b>A</b>) Immunostaining of AQP4 (red) on 12 µm cryosections. Images taken at ×10 and ×0.5 magnifications (inserts: ×20 and ×1 magnifications). The white arrows indicate labeled vessels (scale bar: 100 µm). (<b>B</b>) Immunostaining of α1-syntrophin (red) on 12 µm cryosections. Images were taken at ×10 and ×0.5 magnifications (inserts: ×20 and ×1 magnifications). The white arrows indicate labeled vessels (scale bar: 100 µm).</p>
Full article ">Figure 4
<p>Dp71 re-expression following intracerebroventricular injections (ICVs). Mice were injected at 7 weeks with the AAV9-CAG-GFP control vector (WT-C, Dp71-C) and with the AAV9-CBA-GFP-2A-Dp71 vector (Dp71-T). (<b>A</b>) Transduction territories of the AAV9-CAG-GFP vector in the WT and Dp71-null mouse brain sections, as revealed by GFP immunofluorescent staining (green) (scale bar: 100 µm). (<b>B</b>) Dp71 protein levels in the hippocampus (HIP), cortex (CX) and cerebellum (CBL) quantified by Western blots. The image on the left shows the Western blot for the 6 treated mice (DP-T) and one WT control mice (WT-C). Dp71 was detected using the H4 antibody. Vinculin (Vinc) was used as the control protein for normalization. Importantly, note that all (n = 6) treated mice showed Dp71 protein rescue. (<b>C</b>) Dp71 expression revealed by immunofluorescent staining with the pan-specific H4 antibody (red) on 12 µm cryosections of hippocampus and cerebellum. Images were taken at ×10 and ×0.5 magnifications (inserts: ×20 and ×1 magnifications) (scale bars: 100 µm). SO: stratum oriens; SP: <span class="html-italic">stratum pyramidale</span>; SR: <span class="html-italic">stratum radiatum</span>; MCL: molecular cell layer.</p>
Full article ">Figure 5
<p>Expressions of AQP4 and α1-syntrophin along the walls of blood vessels after intracerebroventricular administration (ICV) of the AAV vectors. (<b>A</b>,<b>B</b>) For both antibodies the immunofluorescent (IF) staining is shown in tissue sections of the hippocampus and cerebellum, as indicated, in the Dp71-null mice injected with the AAV9-CBA-GFP-2A-Dp71 vector (Dp71-T) and with the control vector (Dp71-C, WT, C). (<b>A</b>) Immunostaining of AQP4 (red) on 12 µm cryosections. The white arrows indicate labeled vessels. Images were taken at ×10 and ×0.5 magnifications (inserts: ×20 and ×1 magnifications) (scale bar: 100 µm). (<b>B</b>) Immunostaining of α-1-syntrophin (red) on 12 µm cryosections. The white arrows indicate labeled vessels. Images were taken at ×20 and ×1 magnifications (scale bar: 50 µm).</p>
Full article ">Figure 6
<p>Behavioral study following ICP4 and ICV administration of the AAV vectors. (<b>A</b>–<b>C</b>) The plots show the data recorded following intracardiac injections at P4 (ICP4) in the three groups of mice (Dp71-T, n = 7; WT-C, n = 9; Dp71-C, n = 7). (<b>A</b>) Latency to the first entry in the lit box (s), number of entries and time spent in the lit box (s) in the light–dark choice test (LD). (<b>B</b>) Total number of arms visited, time spent (%) and number of entries (%) in the open arms in the elevated plus maze (EPM). (<b>C</b>) Distance traveled (m), average speed (m/s) and percent distance traveled in center in the 30 min open field exploration test (OF). (<b>D</b>–<b>F</b>) The plots show the data recorded following stereotaxic intracerebroventricular (ICV) injections at 6–8 weeks in the three groups of mice (Dp71-T, n = 6; WT-C, n = 6; Dp71-C, n = 6). (<b>D</b>) Latency to the first entry in the lit box (s), number of entries and time spent in the lit box (s) in the light–dark choice test (LD). (<b>E</b>) Total number of arms visited, time spent (%) and number of entries (%) in the open arms in the elevated plus maze (EPM). (<b>F</b>) Distance traveled (m), average speed (m/s) and percent distance traveled in center in the 30 min open field exploration test (OF). The results are the means ± SEM, * <span class="html-italic">p</span> &lt; 0.05, ns for non-significant, Mann–Whitney test.</p>
Full article ">
12 pages, 3156 KiB  
Article
Development and Characterization of a Novel FVB-PrkdcR2140C Mouse Model for Adriamycin-Induced Nephropathy
by Masaki Watanabe, Yuki Ishii, Kazuki Hashimoto, Hayato R. Takimoto and Nobuya Sasaki
Genes 2024, 15(4), 456; https://doi.org/10.3390/genes15040456 - 4 Apr 2024
Viewed by 852
Abstract
The Adriamycin (ADR) nephropathy model, which induces podocyte injury, is limited to certain mouse strains due to genetic susceptibilities, such as the PrkdcR2140C polymorphism. The FVB/N strain without the R2140C mutation resists ADR nephropathy. Meanwhile, a detailed analysis of the progression of [...] Read more.
The Adriamycin (ADR) nephropathy model, which induces podocyte injury, is limited to certain mouse strains due to genetic susceptibilities, such as the PrkdcR2140C polymorphism. The FVB/N strain without the R2140C mutation resists ADR nephropathy. Meanwhile, a detailed analysis of the progression of ADR nephropathy in the FVB/N strain has yet to be conducted. Our research aimed to create a novel mouse model, the FVB-PrkdcR2140C, by introducing PrkdcR2140C into the FVB/NJcl (FVB) strain. Our study showed that FVB-PrkdcR2140C mice developed severe renal damage when exposed to ADR, as evidenced by significant albuminuria and tubular injury, exceeding the levels observed in C57BL/6J (B6)-PrkdcR2140C. This indicates that the FVB/N genetic background, in combination with the R2140C mutation, strongly predisposes mice to ADR nephropathy, highlighting the influence of genetic background on disease susceptibility. Using RNA sequencing and subsequent analysis, we identified several genes whose expression is altered in response to ADR nephropathy. In particular, Mmp7, Mmp10, and Mmp12 were highlighted for their differential expression between strains and their potential role in influencing the severity of kidney damage. Further genetic analysis should lead to identifying ADR nephropathy modifier gene(s), aiding in early diagnosis and providing novel approaches to kidney disease treatment and prevention. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Urinary albumin excretion in Adriamycin (ADR)-treated B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (B6), BALB/c (BALB), and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (FVB) mice. (<b>a</b>) SDS-PAGE analysis of representative individual B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span>, BALB/c, and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> mice. Bovine serum albumin: BSA (<b>b</b>) Urinary albumin excretion in control B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 5), BALB/c (n = 4), and FVB- <span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 7), as well as in B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> mice (n = 8), BALB/c mice (n = 5), and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> mice (n = 8), 7 days after ADR administration. (<b>c</b>) Urinary albumin excretion in control B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 6), BALB/c (n = 4), and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 5) mice and in ADR-treated B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 8), BALB/c (n = 5), and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 5) mice, 28 days after ADR administration. A vs. B: <span class="html-italic">p</span> &lt; 0.05. A vs. C: <span class="html-italic">p</span> &lt; 0.05. A vs. BC: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Histological analysis of kidneys from Adriamycin (ADR)-treated mice. (<b>a</b>) Representative periodic acid-Schiff-stained renal sections from ADR-treated mice. B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: B6, BALB/c: BALB, FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: FVB. (<b>b</b>) Tubular height. ADR-treated B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 4), BALB/c (n = 4), and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 4) mice were analyzed. A vs. B: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Histological <a href="#genes-15-00456-f003" class="html-fig">Figure 3</a>. Interstitial fibrosis with picrosirius red staining. (<b>a</b>) Representative picrosirius-red-stained images of kidney sections from Adriamycin (ADR)-treated mice. B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: B6, BALB/c: BALB, FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: FVB. (<b>b</b>) Fibrosis area vs. total area. ADR-treated B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 4), BALB/c (n = 4), and FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span> (n = 4) mice were analyzed. A vs. B: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>RNA sequencing analysis. (<b>a</b>) PCA analysis. (<b>b</b>) Heatmap of clustering analysis. (<b>c</b>) Overview of kidney disease resistance genes between C57BL/6 and BALB. (<b>d</b>) Heatmap of differentially expressed genes in the resistance gene locus. Analyses were performed with n = 1 for each strain. B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: B6, BALB/c: BALB, FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: FVB.</p>
Full article ">Figure 5
<p>Expression analysis of differentially expressed genes in the resistance locus after administration of Adriamycin (ADR). Analyses were performed with n = 5 for each strain. B6-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: B6, BALB/c: BALB, FVB-<span class="html-italic">Prkdc<sup>R2140C</sup></span>: FVB. A vs. B: <span class="html-italic">p</span> &lt; 0.05. A vs. C: <span class="html-italic">p</span> &lt; 0.05. B vs. C: <span class="html-italic">p</span> &lt; 0.05. A vs. BC: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
20 pages, 21642 KiB  
Article
Anatomical Anal Stenosis after PPH: Insights from a Retrospective Study and Rat Model
by Chia-Cheng Wen, Shih-Ming Huang and Yi-Wen Wang
Int. J. Mol. Sci. 2024, 25(6), 3543; https://doi.org/10.3390/ijms25063543 - 21 Mar 2024
Viewed by 925
Abstract
High-grade hemorrhoids are usually recommended to receive operational treatments. However, these traditional surgeries are associated with severe postoperative pain. A procedure for prolapse and hemorrhoids (PPH), a circular staple device, has been developed to improve short-term outcomes, including reducing the severity of postoperative [...] Read more.
High-grade hemorrhoids are usually recommended to receive operational treatments. However, these traditional surgeries are associated with severe postoperative pain. A procedure for prolapse and hemorrhoids (PPH), a circular staple device, has been developed to improve short-term outcomes, including reducing the severity of postoperative pain. PPH, compared to conventional surgery, has been associated with the incidence of anatomical anal stenosis. The causes of stenosis after PPH are not yet clear. We first analyzed the complications of our patients with PPH, and then developed a rat model to verify the tension force of PPH using Hematoxylin-eosin, Masson’s trichrome, immunohistochemistry, and immunofluorescence staining. Our clinical data showed that PPH significantly improved postoperative pain, but that it resulted in higher incidences of complications, including anal stenosis, than hemorrhoidectomy. We simulated the status of PPH and developed a rat model to verify PPH’s tension force, including the scarring area and the deposition of proinflammatory factors, angiogenic factors, and fibrotic factors. The tension wound histological data showed more extensive granulation tissue and inflammatory cell infiltration and a thicker epidermis than the control group on day 12 post-operation and tension treatment. In addition to IL-1β and IL-10 cytokines on day 3 and IL-1β, IL-6, and IL-10 cytokines on day 12 post-operation in the tension group, two angiogenic factors, CD31 and VEGF-A, were found to have a more significant expression on day 7 post-operation in the tension group. The mean scar area was larger and the distribution of fibrotic proteins (collagen 1, α-SMA, CTGF, and MMP2) in the tension group was significantly broader than in the control on day 12 post-operation and tension treatment. Based on the findings of our animal model, the development of a lesser tensile force for PPH to decrease the deposition of proinflammatory factors, angiogenic factors, and fibrotic factors is urgently required. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Carton schematic of the anal clock to demonstrate how to define the wound range ratio in anus surgery and the progression of anal stenosis after the PPH surgery. (<b>A</b>) The normal condition of the anus; (<b>B</b>) the open (Milligan–Morgan) hemorrhoidectomy; (<b>C</b>) the closed (Ferguson) hemorrhoidectomy. θ represents the angle of the surgical wound extent in a column. (<b>D</b>) Initially, after the PPH surgery, the wound showed normal wound tension. (<b>E</b>) During wound healing, the tension of the ring wound increased slightly toward the inner anus, and then a slight scar was formed, while the patient’s defecation function was not affected. (<b>F</b>) Finally, as the wound healing gradually progressed, the wound tension continued to increase, leading to hypertrophic scars, and finally severe anal stenosis.</p>
Full article ">Figure 2
<p>The tool and quantifiable tension applied in this study. (<b>A</b>) Picture of the spreader, the distance of both ends was 2.5 cm in a relaxed state. (<b>B</b>) In the tension group, the spreader pressed both ends to a distance of 2 cm and was fixed to the incision wound, producing a tension of about 85 gm. In the control group, the spreader was fixed to the incision wound without pressing both ends. (<b>C</b>) The appearance of linear wound healing progress from day 0 (D0) to day 12 (D12) in the tension and control groups.</p>
Full article ">Figure 3
<p>Histological images of inflammatory cell infiltration in the control and tension groups. (<b>A</b>) H&amp;E staining, quantitative data of the inflammatory cell infiltration area. Data are presented as the mean ± standard deviation (n = 8; * <span class="html-italic">p</span> &lt; 0.05). (<b>B</b>,<b>C</b>) M1 and M2 cell distribution of the granulation tissue on days 3, 7, and 12. The data of (<b>C</b>) were further analyzed from the (<b>B</b>) D12 data (red lines for control group and yellow lines for tension group).</p>
Full article ">Figure 4
<p>Inflammatory-related cytokines in the control and tension groups. Immunohistochemistry staining and relative quantitative data of IL-1β, IL-6, IL-10, and TGF-β1 in the control and tension groups on day 3 (<b>A</b>,<b>C</b>) and day 12 (<b>B</b>,<b>D</b>) after the operation. Scale bar = 400 μm (<b>D</b>) and 500 μm. Data of integrated density of staining within a defined area (IntDen/Area) are presented as the mean ± standard deviation (n = 3 at D3, n = 8 at D12; * <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; 0.0001).</p>
Full article ">Figure 5
<p>Immunohistochemistry staining and relative quantitative data of CD31 and VEGF-A in the tension and control groups on day 7 after the operation. Scale bar = 400 μm. Data of integrated density of staining within a defined area (IntDen/Area) are presented as the mean ± standard deviation (n = 3; * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Histological analysis of scar tissue from the tension and control groups on day 12 after the operation. (<b>A</b>) Masson’s trichrome staining, (<b>B</b>) the scar area, (<b>C</b>) deposition percentage of collagen fiber quantitative data (% of area), (<b>D</b>,<b>E</b>) immunohistochemistry staining and relative quantitative data of type one collagen. Data of integrated density of staining within a defined area (IntDen/Area) are presented as the mean ± standard deviation (n = 8; ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 7
<p>Immunohistochemistry staining and relative quantitative data of (<b>A</b>,<b>B</b>) α-SMA, (<b>C</b>,<b>D</b>) CTCF, (<b>E</b>,<b>F</b>) cyr61, (<b>G</b>,<b>H</b>) MMP-2, and (<b>I</b>,<b>J</b>) MMP-9 in the control and tension groups on day 12 after the operation. Data are presented as the mean ± standard deviation (n = 8; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, NS: not significant).</p>
Full article ">
15 pages, 1996 KiB  
Article
Therapeutic Effects of a Novel Aptamer on Coronaviral Infection-Induced Lung Injury and Systemic Inflammatory Responses
by Yingchun Wang, Mikael Lindstam, David Hwang, Luiza Jedlina and Mingyao Liu
Cells 2024, 13(5), 422; https://doi.org/10.3390/cells13050422 - 28 Feb 2024
Viewed by 1337
Abstract
Background: Coronaviral infection-induced acute lung injury has become a major threat to public health, especially through the ongoing pandemic of COVID-19. Apta-1 is a newly discovered Aptamer that has anti-inflammatory effects on systemic septic responses. The therapeutic effects of Apta-1 on coronaviral infection-induced [...] Read more.
Background: Coronaviral infection-induced acute lung injury has become a major threat to public health, especially through the ongoing pandemic of COVID-19. Apta-1 is a newly discovered Aptamer that has anti-inflammatory effects on systemic septic responses. The therapeutic effects of Apta-1 on coronaviral infection-induced acute lung injury and systemic responses were evaluated in the present study. Methods: Female A/J mice (at 12–14 weeks of age) were challenged with murine hepatitis virus 1 (MHV-1), a coronavirus, at 5000 PFU intranasally, followed by Apta-1 intravenously administered (100 mg/kg, twice) 1.5 h or 2 days after viral delivery. Animals were sacrificed at Day 2 or Day 4. Lung tissues were examined with H&E, immunohistochemistry staining, and western blotting. RT-qPCR was used for cytokine gene expression. Serum and plasma were collected for laboratory assessments. Results: Apta-1 treatment reduced viral titers, prevented MHV-1-induced reduction of circulating blood volume and hemolysis, reduced alveolar space hemorrhage, and protease-activated receptor 1 (PAR-1) cleavage. Apta-1 treatment also significantly reduced chemokine (MKC, MCP-1, and RANTES) levels, as well as AST, ALT, total bilirubin, and reduced unconjugated bilirubin levels in the serum. Conclusion: Apta-1 showed therapeutic benefits in coronaviral infection-induced hemorrhage and PAR-1 cleavage in the lung. It also has anti-inflammatory effects systemically. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Apta-1 inhibits LPS-induced acute inflammatory responses in mouse lungs. (<b>A</b>). Experimental design. Female C57BL/6J mice were challenged intraperitoneally with LPS (10 mg/kg), followed by an intravenous injection of Apta-1 (100 mg/kg, twice). One day later, animals were sacrificed, and gene expression of cytokines in the lung was analyzed using RT-qPCR. Apta-1 treatment significantly inhibited LPS-induced gene expression of <span class="html-italic">Ip-10</span> (<b>B</b>), <span class="html-italic">Ptx3</span> (<b>C</b>), <span class="html-italic">Il-6</span> (<b>D</b>), and reduced the expression of <span class="html-italic">Mip-1</span> (<b>E</b>), <span class="html-italic">Il-10</span> (<b>F</b>), and <span class="html-italic">Il-12a</span> (<b>G</b>), although it did not reach statistical significance. Expression of the gene of interest was normalized with housekeeping gene <span class="html-italic">Ppi1b</span> and expressed as the ratio with the average of the control group. *: <span class="html-italic">p</span> &lt; 0.05. One-way ANOVA followed by the unpaired <span class="html-italic">t</span>-test. The number of mice: 4 for control, 7 for LPS, and 8 for LPS plus Apta-1.</p>
Full article ">Figure 2
<p>Apta-1 reduces the MHV-1 coronaviral titer, viral infection-induced loss of circulating blood, and hemolysis. (<b>A</b>). Dose responses of MHV-1 on body weight changes in mice. Female A/J mice at 12-14 weeks of age were challenged intratracheally with the MHV-1 coronavirus at 500 PFU (n = 6), 1000 PFU (n = 6), or 5000 PFU (n = 3). At 5000 PFU, MHV-1 induced more body weight lose with delayed recovery. This dose was used in the subsequent studies. (<b>B</b>). Experimental design. After intranasal delivery of the MHV-1 coronavirus, A/J mice received the Apta-1 treatment (100 mg/kg, twice) at Day 2 and were sacrificed at Day 4 (D2D4) or received the Apta-1 at Day 0 and were sacrificed at Day 2 (D0D2) or Day 4 (D0D4). Mice in control groups received an equal volume of vehicle solution. (<b>C</b>). The MHV-1 viral titer was significantly lower at Day 4 (<span class="html-italic">p</span> &lt; 0.0001). Apta-1 treatment significantly reduced the viral titer in the D2D4 group. (<b>D</b>). The MHV-1 viral infection induced more body weight loss at Day 4 (<span class="html-italic">p</span> &lt; 0.0001). Apta-1 did not affect the MHV-1 induced body weight changes. (<b>E</b>). The serum volumes collected from the D0D2 and D0D4 groups were significantly higher in the Apta-1 treated animals (<span class="html-italic">t</span>-test). (<b>F</b>). The MHV-1 viral infection induced more hemolysis at Day 4 (<span class="html-italic">p</span> &lt; 0.0001). The Apta-1 treatment significantly reduced hemolysis in the D0D4 group. Two-way ANOVA followed by the Mann–Whitney test. *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01. Numbers of mice in MHV-1 and MHV-1 + Apta-1 groups are: 7 and 8 in D0D2 groups; 8 and 8 in D0D4 groups; 9 and 9 in D2D4 groups.</p>
Full article ">Figure 3
<p>Apta-1 treatment reduced alveolar space hemorrhage. The lung injury score was blindly evaluated by a pathologist. (<b>A</b>). Alveolar space hemorrhage was significantly less in the Apta-1 treated groups than in the MHV-1 only groups. The vertical line highlights the two experimental conditions. *: <span class="html-italic">p</span> &lt; 0.05; two-way ANOVA. Numbers of mice in MHV-1 and MHV-1 + Apta-1 groups are: 7 and 8 in D0D2 groups; 8 and 7 in D0D4 groups; 9 and 8 in D2D4 groups. (<b>B</b>) and (<b>C</b>). D0D2 groups were repeated with inflation perfusion fixation. (<b>B</b>). Alveolar space hemorrhage (indicated with arrows) is seen in the MHV-1 group. The scale bar is 200 μm. (<b>C</b>). The Apta-1 treatment prevented alveolar space hemorrhage (one-way ANOVA followed by the Mann–Whitney test). Numbers of mice in control, MHV-1, and MHV-1 + Apta-1 groups are: 5, 15, and 15, respectively.</p>
Full article ">Figure 4
<p>Apta-1 treatment significantly reduced PAR-1 cleavage in the lung tissue of MHV-1 viral infected mice. (<b>A</b>). Two sets of original western blots as examples show two bands of PAR-1 proteins. In the MHV-1 viral infected lung tissue, the lower bands became dominate and the top bands were shifted towards the lower bands. NA: no administration; CTL: control animals inoculated with DMEM instead of MHV-1 as a vehicle control and then treated with saline instead of Apta-1. (<b>B</b>). In inflated perfused lung tissue, the ratio of lower bands to higher bands was significantly lower in the Apta-1 treated lungs (<span class="html-italic">t</span>-test). Numbers of mice in MHV-1 and MHV-1 + Apta-1 groups are: 13 and 15, respectively. (<b>C</b>). In non-perfused lungs, the ratio was significantly increased at Day 4, and it was significantly lower in the Apta-1 treated groups (two-way ANOVA). The vertical line highlights the two experimental conditions. *: <span class="html-italic">p</span> &lt; 0.05. Numbers of mice in MHV-1 and MHV-1 + Apta-1 groups are: 5 and 6 in D0D2 groups; 7 and 8 in D0D4 groups; 8 and 8 in D2D4 groups. Equal loading was confirmed with GAPDH as a housekeeping protein.</p>
Full article ">Figure 5
<p>Apta-1 treatment significantly reduce several cytokine levels in the serum. MHV-1 airway infection-induced circulating levels of IL-6 (<b>A</b>), IFN-γ (<b>B</b>) and IP-10 (<b>C</b>) were significantly reduced at Day 4, which were not affected by Apta-1 treatment. The serum levels of MKC (<b>D</b>), MCP-1 (<b>E</b>), and RANTES (<b>F</b>), were significantly lower in Apta-1 treated animals. Two-way ANOVA. Vertical line highlights two experimental conditions. *: <span class="html-italic">p</span> &lt; 0.05. Numbers of mice in MHV-1 and MHV-1 + Apta-1 groups are: 7 and 8 in D0D2 groups; 8 and 8 in D0D4 groups; 9 and 9 in D2D4 groups.</p>
Full article ">Figure 6
<p>The Apta-1 treatment significantly reduced AST, total bilirubin, and unconjugated bilirubin in the serum. The MHV-1 airway infection induced a significant increase in AST (<b>A</b>), ALT (<b>B</b>), total bilirubin (<b>C</b>), and unconjugated bilirubin (<b>D</b>) in the serum at Day 4. The Apta-1 treated animals showed significantly lower levels of AST in the D0D4 group, and significantly lower levels of total bilirubin and unconjugated bilirubin. Two-way ANOVA. The vertical line highlights the two experimental conditions. *: <span class="html-italic">p</span> &lt; 0.05. Numbers of mice in MHV-1 and MHV-1 + Apta-1 groups are: 7 and 8 in D0D2 groups; 8 and 8 in D0D4 groups; 9 and 9 in D2D4 groups.</p>
Full article ">
30 pages, 13237 KiB  
Article
Effects of Paraquat, Dextran Sulfate Sodium, and Irradiation on Behavioral and Cognitive Performance and the Gut Microbiome in A53T and A53T-L444P Mice
by Ariel Chaklai, Abigail O’Neil, Shrey Goel, Nick Margolies, Destine Krenik, Ruby Perez, Kat Kessler, Elizabeth Staltontall, Hong Ki (Eric) Yoon, Montzerrat Pantoja, Keaton Stagaman, Kristin Kasschau, Vivek Unni, Robert Duvoisin, Thomas Sharpton and Jacob Raber
Genes 2024, 15(3), 282; https://doi.org/10.3390/genes15030282 - 23 Feb 2024
Viewed by 1566
Abstract
Heterozygous carriers of the glucocerebrosidase 1 (GBA) L444P Gaucher mutation have an increased risk of developing Parkinson’s disease (PD). The GBA mutations result in elevated alpha synuclein (aSyn) levels. Heterozygous mice carrying one allele with the L444P mutation knocked-into the mouse gene show [...] Read more.
Heterozygous carriers of the glucocerebrosidase 1 (GBA) L444P Gaucher mutation have an increased risk of developing Parkinson’s disease (PD). The GBA mutations result in elevated alpha synuclein (aSyn) levels. Heterozygous mice carrying one allele with the L444P mutation knocked-into the mouse gene show increased aSyn levels and are more sensitive to motor deficits following exposure to the neurotoxin (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) MPTP than wild-type mice. Paraquat (PQ), a herbicide, increases PD risk in most studies. Its effects on the brain involve alterations in the gut microbiome. Exposure to dextran sulfate sodium (DSS), a mouse model of colitis, can be used to determine whether gut microbiome alterations are sufficient to induce PD-relevant phenotypes. We rederived the A53T-L444P and A53T mouse lines to assess whether PQ, PQ in combination with radiation exposure (IR), and DSS have differential effects in A53T and A53T-L444P mice and whether these effects are associated with alterations in the gut microbiome. PQ and PQ + IR have differential effects in A53T and A53T-L444P mice. In contrast, effects of DSS are only seen in A53T-L444P mice. Exposure and genotype modulate the relationship between the gut microbiome and behavioral performance. The gut microbiome may be an important mediator of how environmental exposures or genetic mutations yield behavioral and cognitive impacts. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Schematic outline of study 1.</p>
Full article ">Figure 2
<p>Schematic outline of study 2.</p>
Full article ">Figure 3
<p>Schematic outline of study 3.</p>
Full article ">Figure 4
<p>Schematic outline of study 4.</p>
Full article ">Figure 5
<p>(<b>A</b>) For all figures, in cases that there was a main effect of sex or a statistical interaction with sex, female and male data were analyzed separately. All groups showed spatial habituation learning and moved less in the open field over the three subsequent days (<span class="html-italic">F</span> (1,75) = 84.686, <span class="html-italic">p</span> &lt; 0.001). As there was a treatment × sex interaction (<span class="html-italic">F</span> (1,75) = 6.152, <span class="html-italic">p</span> = 0.015), we also analyzed the male and female data separately. (<b>B</b>) In males, there was an effect of day (<span class="html-italic">F</span> (1,42) = 93.550, <span class="html-italic">p</span> &lt; 0.001) and an effect of treatment (<span class="html-italic">F</span> (1,42) = 93.550, *** <span class="html-italic">p</span> &lt; 0.001). PQ-treated mice moved less than saline-treated mice. (<b>C</b>) In females, there was only an effect of day (<span class="html-italic">F</span> (2,66) = 15.320, <span class="html-italic">p</span> &lt; 0.001). (<b>D</b>) For time spent in the more anxiety-provoking center of the open field, there was an effect of day (<span class="html-italic">F</span> (1,4) = 42.179, <span class="html-italic">p</span> &lt; 0.001) and an effect of treatment (<span class="html-italic">F</span> (1,74) = 4.205, <span class="html-italic">p</span> = 0.044). When only the day 1 data were analyzed, there was an effect of treatment, with PQ-treated mice spending less time in the center of the open field on day 1 (<span class="html-italic">F</span> (1,74) = 6.091, * <span class="html-italic">p</span> = 0.016). (<b>E</b>) In the two days of the open field containing objects, there was no effect of genotype or treatment. There was no preferential exploration of the novel object in any of the groups. (<b>F</b>) For time spent exploring the objects, there was an effect of treatment (<span class="html-italic">F</span> (1,75) = 4.809, <span class="html-italic">p</span> = 0.031) and a treatment × genotype interaction (<span class="html-italic">F</span> (1,74) = 7.098, <span class="html-italic">p</span> = 0.009). When only A53T mice were analyzed, there was an effect of treatment (<span class="html-italic">F</span> (1,33) = 7.619, ** <span class="html-italic">p</span> = 0.009). PQ-treated A53T mice spent less time exploring the objects than saline-treated A53T mice. (<b>G</b>) For the discrimination index, there were no effects of genotype or treatment. None of the groups showed a discrimination index different from 0 and therefore no preferential exploration of the novel object. (<b>H</b>) When sensorimotor function was assessed in the rotorod test, there was only an effect of day (<span class="html-italic">F</span>(1,75) = 65.927, *** <span class="html-italic">p</span> &lt; 0.001), with better performance on day 2 than day 1. Circles: A53T; squares: A53T-L444P. Open symbols: saline; filled symbols: PQ.</p>
Full article ">Figure 6
<p>(<b>A</b>) For spontaneous alternation in the Y-maze, there a treatment × sex interaction (<span class="html-italic">F</span>(1,74) = 4.975, <span class="html-italic">p</span> = 0.029). (<b>B</b>) In males, there was a treatment × genotype interaction (<span class="html-italic">F</span>(1,32) = 5.578, <span class="html-italic">p</span> = 0.024). When each genotype was analyzed separately, there was no effect of treatment in either. (<b>C</b>) In females, there was an effect of treatment (<span class="html-italic">F</span>(1,15) = 7.602, * <span class="html-italic">p</span> = 0.015). This effect seemed driven by a higher percent spontaneous alternation in PQ- than saline-treated A53T-L444P mice. (<b>D</b>) For arm entries in the Y-maze, there was a treatment × genotype interaction (<span class="html-italic">F</span>(1,74) = 10.681, <span class="html-italic">p</span> = 0.002). In A53T mice, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,33) = 3.842, <sup>#</sup> <span class="html-italic">p</span> = 0.058). In A53T-L444P mice, there was an effect of treatment (<span class="html-italic">F</span>(1,41) = 7.224, * <span class="html-italic">p</span> = 0.010). PQ-treated A53T-L444P mice were more active than saline-treated A53T-L444P mice. (<b>E</b>) There was no effect of genotype or treatment in depressive-like behavior in the forced swim test but there was a sex × genotype interaction (<span class="html-italic">F</span>(1,74) = 8.111, <span class="html-italic">p</span> = 0.006). (<b>F</b>) There was an effect of genotype in males (<span class="html-italic">F</span>(1,42) = 7.836, ** <span class="html-italic">p</span> = 0.0077). There was more depressive-like behavior in A53T-L444P than A53T mice. (<b>G</b>) There was no effect of genotype in females. Circles: A53T; squares: A53T-L444P. Open symbols: saline; filled symbols: PQ.</p>
Full article ">Figure 7
<p>(<b>A</b>) During the baseline period on the training day, there was a trend towards higher freezing levels in PQ- than saline-treated mice (<span class="html-italic">F</span>(1,75) = 2.856, <sup>#</sup> <span class="html-italic">p</span> = 0.095) on the percent freezing. (<b>B</b>) When activity levels during the baseline period on the training day were analyzed, there was a treatment × sex interaction (<span class="html-italic">F</span>(1,75) = 5.475, <span class="html-italic">p</span> = 0.022). (<b>C</b>) In males, there was an effect of treatment (<span class="html-italic">F</span>(1,42) = 4.928, * <span class="html-italic">p</span> = 0.032). Activity levels were lower in PQ- than saline-treated mice. (<b>D</b>) In females, there was no effect of treatment. (<b>E</b>) There was no effect of genotype or treatment on the freezing levels during the ISI. (<b>F</b>) During the ISI, there was a treatment × sex interaction (<span class="html-italic">F</span>(1,75) = 6.853, <span class="html-italic">p</span> = 0.011) and a genotype × sex interaction (<span class="html-italic">F</span>(1,75) = 5.956, <span class="html-italic">p</span> = 0.017). (<b>G</b>) In males, there were no significant effects or trend towards treatment effects. (<b>H</b>) In A53T-L444P female mice, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,16) = 4.408, <sup>#</sup> <span class="html-italic">p</span> = 0.052). (<b>I</b>) When freezing during the tones was analyzed, there was a tone × sex × genotype × treatment interaction (<span class="html-italic">F</span>(1,75) = 6.612, <span class="html-italic">p</span> = 0.012). In A53T mice, there was only an effect of tone (<span class="html-italic">F</span>(1,33) = 83.234, <span class="html-italic">p</span> &lt; 0.001). In A53T-L444P mice, there was a tone × sex × treatment interaction (<span class="html-italic">F</span>(1,42) = 7.478, <span class="html-italic">p</span> = 0.009) and a tone × treatment interaction (<span class="html-italic">F</span>(1,75) = 5.093, <span class="html-italic">p</span> = 0.029). In A53T-L444P mice during tone 2, there was a treatment × sex interaction (<span class="html-italic">F</span>(1,42) = 6.160, <span class="html-italic">p</span> = 0.017). (<b>J</b>) In A53T-L444P male mice, there was no effect on percent freezing during tone 2. (<b>K</b>) In A53T-L444P female mice, there was an effect of treatment in females (<span class="html-italic">F</span>(1,16) = 5.674, * <span class="html-italic">p</span> = 0.030). Freezing levels were lower in PQ- than saline-treated mice. (<b>L</b>) When activity levels during the tones were analyzed, there was a treatment × sex interaction (<span class="html-italic">F</span>(1,75) = 4.199, <span class="html-italic">p</span> = 0.044). (<b>M</b>) There was no effect of treatment or trend towards an effect of treatment on activity levels during the tones. (<b>N</b>) In females, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,33) = 3.067, <sup>#</sup> <span class="html-italic">p</span> = 0.089) on activity levels during the tones. (<b>O</b>) There was no effect of genotype or treatment on responsiveness to the shocks. Circles: A53T; squares: A53T-L444P. Open symbols: saline; filled symbols: PQ.</p>
Full article ">Figure 8
<p>(<b>A</b>) There was an effect of genotype on percent freezing in the contextual fear memory test (<span class="html-italic">F</span>(1,71) = 4.448, * <span class="html-italic">p</span> = 0.038). Contextual fear memory was stronger in A53T-L444P than A53T mice. There was also a genotype × treatment interaction (<span class="html-italic">F</span>(1,71) = 5.573, <span class="html-italic">p</span> = 0.021), a genotype × sex interaction (<span class="html-italic">F</span>(1,71) = 5.597, <span class="html-italic">p</span> = 0.021), a treatment × sex interaction (<span class="html-italic">F</span>(1,71) = 4.178, <span class="html-italic">p</span> = 0.045), and a genotype × treatment × sex interaction (<span class="html-italic">F</span>(1,71) = 10.463, <span class="html-italic">p</span> = 0.002). (<b>B</b>) In A53T-L444P male mice, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,25) = 3.866, <sup>#</sup> <span class="html-italic">p</span> = 0.060). (<b>C</b>) In A53T-L444P female mice, there was an effect of treatment (<span class="html-italic">F</span>(1,75) = 14.725, * <span class="html-italic">p</span> = 0.002), with reduced freezing in PQ-treated female mice. ** <span class="html-italic">p</span> &lt; 0.01. (<b>D</b>) When activity levels were analyzed in the contextual fear memory test, there was a genotype × treatment interaction (<span class="html-italic">F</span>(1,71) = 6.067, <span class="html-italic">p</span> = 0.016), a genotype × sex interaction (<span class="html-italic">F</span>(1,75) = 8.423, <span class="html-italic">p</span> = 0.005), a treatment × sex interaction (<span class="html-italic">F</span>(1,71) = 6.307, <span class="html-italic">p</span> = 0.014), and a genotype × treatment × sex interaction (<span class="html-italic">F</span>(1,75) = 7.482, <span class="html-italic">p</span> = 0.008). (<b>E</b>) In A53T-L444P males, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,25) = 3.263, <sup>#</sup> <span class="html-italic">p</span> = 0.083). (<b>F</b>) In A53T-L444P females, there was an effect of treatment (<span class="html-italic">F</span>(1,15) = 8.201, * <span class="html-italic">p</span> = 0.012). Activity levels were higher in PQ- than saline-treated mice. (<b>G</b>) In the cued fear memory test, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 9.391, * <span class="html-italic">p</span> = 0.003), with higher freezing levels in A53T-L444P than A53T mice. There was also a period × genotype interaction (<span class="html-italic">F</span>(1,75) = 6.537, <span class="html-italic">p</span> = 0.013). During the pre-tone period, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 4.381, <span class="html-italic">p</span> = 0.040), with higher freezing levels in A53T-L444P mice, and treatment (<span class="html-italic">F</span>(1,75) = 4.353, <sup>o</sup> <span class="html-italic">p</span> = 0.040), with higher freezing levels in PQ- than saline-treated mice. When percent freezing was analyzed during the tone, there was only an effect of genotype (<span class="html-italic">F</span>(1,75) = 8.885, <span class="html-italic">p</span> = 0.004), with higher freezing in A53T-L444P than A53T mice. (<b>H</b>) When activity levels during the pre-tone and tone periods were analyzed in the cued fear memory test, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 5.871, * <span class="html-italic">p</span> = 0.018), with lower activity levels in A53T-L444P than A53T mice. There was also an effect of period (<span class="html-italic">F</span>(1,75) = 272.918, <span class="html-italic">p</span> &lt; 0.001), with lower activity during the tone than pre-tone period. When activity levels during the tone in the cued fear memory test were analyzed, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 8.667, <span class="html-italic">p</span> = 0.004), with lower activity levels in A53T-L444P than A53T mice. Circles: A53T; squares: A53T-L444P. Open symbols: saline; filled symbols: PQ.</p>
Full article ">Figure 8 Cont.
<p>(<b>A</b>) There was an effect of genotype on percent freezing in the contextual fear memory test (<span class="html-italic">F</span>(1,71) = 4.448, * <span class="html-italic">p</span> = 0.038). Contextual fear memory was stronger in A53T-L444P than A53T mice. There was also a genotype × treatment interaction (<span class="html-italic">F</span>(1,71) = 5.573, <span class="html-italic">p</span> = 0.021), a genotype × sex interaction (<span class="html-italic">F</span>(1,71) = 5.597, <span class="html-italic">p</span> = 0.021), a treatment × sex interaction (<span class="html-italic">F</span>(1,71) = 4.178, <span class="html-italic">p</span> = 0.045), and a genotype × treatment × sex interaction (<span class="html-italic">F</span>(1,71) = 10.463, <span class="html-italic">p</span> = 0.002). (<b>B</b>) In A53T-L444P male mice, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,25) = 3.866, <sup>#</sup> <span class="html-italic">p</span> = 0.060). (<b>C</b>) In A53T-L444P female mice, there was an effect of treatment (<span class="html-italic">F</span>(1,75) = 14.725, * <span class="html-italic">p</span> = 0.002), with reduced freezing in PQ-treated female mice. ** <span class="html-italic">p</span> &lt; 0.01. (<b>D</b>) When activity levels were analyzed in the contextual fear memory test, there was a genotype × treatment interaction (<span class="html-italic">F</span>(1,71) = 6.067, <span class="html-italic">p</span> = 0.016), a genotype × sex interaction (<span class="html-italic">F</span>(1,75) = 8.423, <span class="html-italic">p</span> = 0.005), a treatment × sex interaction (<span class="html-italic">F</span>(1,71) = 6.307, <span class="html-italic">p</span> = 0.014), and a genotype × treatment × sex interaction (<span class="html-italic">F</span>(1,75) = 7.482, <span class="html-italic">p</span> = 0.008). (<b>E</b>) In A53T-L444P males, there was a trend towards an effect of treatment (<span class="html-italic">F</span>(1,25) = 3.263, <sup>#</sup> <span class="html-italic">p</span> = 0.083). (<b>F</b>) In A53T-L444P females, there was an effect of treatment (<span class="html-italic">F</span>(1,15) = 8.201, * <span class="html-italic">p</span> = 0.012). Activity levels were higher in PQ- than saline-treated mice. (<b>G</b>) In the cued fear memory test, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 9.391, * <span class="html-italic">p</span> = 0.003), with higher freezing levels in A53T-L444P than A53T mice. There was also a period × genotype interaction (<span class="html-italic">F</span>(1,75) = 6.537, <span class="html-italic">p</span> = 0.013). During the pre-tone period, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 4.381, <span class="html-italic">p</span> = 0.040), with higher freezing levels in A53T-L444P mice, and treatment (<span class="html-italic">F</span>(1,75) = 4.353, <sup>o</sup> <span class="html-italic">p</span> = 0.040), with higher freezing levels in PQ- than saline-treated mice. When percent freezing was analyzed during the tone, there was only an effect of genotype (<span class="html-italic">F</span>(1,75) = 8.885, <span class="html-italic">p</span> = 0.004), with higher freezing in A53T-L444P than A53T mice. (<b>H</b>) When activity levels during the pre-tone and tone periods were analyzed in the cued fear memory test, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 5.871, * <span class="html-italic">p</span> = 0.018), with lower activity levels in A53T-L444P than A53T mice. There was also an effect of period (<span class="html-italic">F</span>(1,75) = 272.918, <span class="html-italic">p</span> &lt; 0.001), with lower activity during the tone than pre-tone period. When activity levels during the tone in the cued fear memory test were analyzed, there was an effect of genotype (<span class="html-italic">F</span>(1,75) = 8.667, <span class="html-italic">p</span> = 0.004), with lower activity levels in A53T-L444P than A53T mice. Circles: A53T; squares: A53T-L444P. Open symbols: saline; filled symbols: PQ.</p>
Full article ">Figure 9
<p>(<b>A</b>) PQ + IR-treated mice moved less than control mice in the open field (<span class="html-italic">F</span>(1,58) = 4.668, * <span class="html-italic">p</span> = 0.035). (<b>B</b>) There was also a genotype × treatment interaction (<span class="html-italic">F</span>(1,58) = 4.784, <span class="html-italic">p</span> = 0.033), and a sex × genotype × treatment interaction (<span class="html-italic">F</span>(1,58) = 5.826, <span class="html-italic">p</span> = 0.019) for time spent in the more anxiety-provoking center of the open field. In A53T-L444P mice, anxiety levels were higher in PQ + IR- than control mice. (<b>C</b>) There was no effect of genotype or treatment on measures of anxiety in the open field in male mice. (<b>D</b>) In A53T-L444P mice, PQ + IR-treated female mice spent less time in the center than control-treated female mice (<span class="html-italic">F</span>(1,23) = 4.825, * <span class="html-italic">p</span> = 0.038). (<b>E</b>) There was no effect of genotype or treatment on activity levels in the open field containing objects. (<b>F</b>) For time spent in the center of the open field where the objects were, there was an effect of sex (<span class="html-italic">F</span>(1,58) = 5.385, <span class="html-italic">p</span> = 0.024), a treatment × genotype interaction (<span class="html-italic">F</span>(1,58) = 4.221, <span class="html-italic">p</span> = 0.044), a day × treatment × sex interaction (<span class="html-italic">F</span>(1,58) = 4.692, <span class="html-italic">p</span> = 0.034), a day × treatment × genotype interaction (<span class="html-italic">F</span>(1,58) = 6.167, <span class="html-italic">p</span> = 0.016), and a day × treatment × sex × genotype interaction (<span class="html-italic">F</span>(1,58) = 7.233, <span class="html-italic">p</span> = 0.009). In A53T-L444P mice, there was a trend towards an effect of treatment over the two days of testing in the open field containing objects (<span class="html-italic">F</span>(1,23) = 3.241, <sup>#</sup> <span class="html-italic">p</span> = 0.085) and an effect of treatment on day 2 (<span class="html-italic">F</span>(1,23) = 5.498, * <span class="html-italic">p</span> = 0.028). (<b>G</b>) A53T-L444P male mice were analyzed, and there was an effect of treatment (<span class="html-italic">F</span>(1,14) = 5.297, <sup>#</sup> <span class="html-italic">p</span> = 0.037). (<b>H</b>) In A53T-L444P female mice, there was a day × treatment interaction (<span class="html-italic">F</span>(1,9) = 6.989, <span class="html-italic">p</span> = 0.027) but not an effect of treatment on either day. (<b>I</b>) There was no effect of genotype or treatment on the discrimination index. (<b>J</b>) There was no effect of genotype or treatment on grip strength. (<b>K</b>) There was a trend towards a higher grip strength per body weight in mice that received PQ + IR grip strength (<span class="html-italic">F</span>(1,58) = 3.329, <sup>#</sup> <span class="html-italic">p</span> = 0.073). (<b>L</b>) In the forced swim test, there was more depressive-like behavior in A53T-L444P than A53T mice (<span class="html-italic">F</span>(1,58) = 4.669, * <span class="html-italic">p</span> = 0.035). Circles: A53T; squares: A53T-L444P. Open symbols: control; filled symbols: PQ + IR.</p>
Full article ">Figure 10
<p>(<b>A</b>) During the baseline period on the training day, freezing levels were higher in the PQ + IR- than control-treated mice (<span class="html-italic">F</span>(1,54) = 9.216, *** <span class="html-italic">p</span> = 0.004). (<b>B</b>) During the baseline period on the training day, activity levels were lower in the PQ + IR- than control-treated mice (<span class="html-italic">F</span>(1,54) = 9.845, *** <span class="html-italic">p</span> = 0.003). (<b>C</b>) There was an effect of treatment (<span class="html-italic">F</span>(1,58) = 11.898, *** <span class="html-italic">p</span> = 0.001) and a treatment × genotype interaction (<span class="html-italic">F</span>(1,58) = 5.944, <span class="html-italic">p</span> = 0.018) for response to the shocks. The response to shock was lower in PQ + IR than control mice. (<b>D</b>) In males, PQ + IR-treated mice showed a lower response to the shocks than control-treated mice (<span class="html-italic">F</span>(1,32) = 23.416, **** <span class="html-italic">p</span> &lt; 0.001). (<b>E</b>) In females, there was no effect of genotype or treatment on response to the shocks. (<b>F</b>) PQ + IR-treated mice froze more during the first tone (<span class="html-italic">F</span>(1,54) = 9.986, *** <span class="html-italic">p</span> = 0.003). (<b>G</b>) PQ + IR-treated mice moved less during the first tone (<span class="html-italic">F</span>(1,54) = 9.275, *** <span class="html-italic">p</span> = 0.004). (<b>H</b>) When freezing during the contextual fear memory test was analyzed, there was a treatment × genotype interaction (<span class="html-italic">F</span>(1,58) = 4.037, <span class="html-italic">p</span> = 0.049). In A53T-L444P mice, there was a trend towards lower freezing levels in IR + PQ -treated than control-treated mice (<span class="html-italic">F</span>(1,23) = 3.780, <sup>#</sup> <span class="html-italic">p</span> = 0.064). (<b>I</b>) When activity levels were analyzed in the cued fear memory test, there was an effect of genotype (<span class="html-italic">F</span>(1,58) = 4.604, * <span class="html-italic">p</span> = 0.036). This effect seemed driven by higher activity levels in PQ + IR-treated A53T-L444P than A53T mice. There were also a treatment × genotype × sex interaction (<span class="html-italic">F</span>(1,58) = 6.048, <span class="html-italic">p</span> = 0.017) and a period × treatment × genotype × sex interaction (<span class="html-italic">F</span>(1,58) = 6.874, <span class="html-italic">p</span> = 0.011). In A53T mice, there was only an effect of period (<span class="html-italic">F</span>(1,35) = 126.536, <span class="html-italic">p</span> &lt; 0.001). In A53T-L444P mice, there was a treatment × sex interaction (<span class="html-italic">F</span>(1,23) = 6.233, <span class="html-italic">p</span> = 0.020), a period × treatment × sex interaction (<span class="html-italic">F</span>(1,23) = 5.265, <span class="html-italic">p</span> = 0.031), and trends towards an effect of sex (<span class="html-italic">F</span>(1,58) = 3.040, <span class="html-italic">p</span> = 0.095) and a period × sex interaction (<span class="html-italic">F</span>(1,58) = 3.914, <span class="html-italic">p</span> = 0.060). During the pre-tone period, activity levels were higher in A53T-L44P than A53T mice (<span class="html-italic">F</span>(1,58) = 4.816, <sup>o</sup> <span class="html-italic">p</span> = 0.032). (<b>J</b>) In males, there was only an effect of period. (<b>K</b>) When activity levels were only analyzed in A53T-L444P females, there was as effect of treatment (<span class="html-italic">F</span>(1,9) = 8.163, * <span class="html-italic">p</span> = 0.019), with lower activity levels in IR + PQ- than control-treated A53T mice and higher activity levels in IR + PQ- than control-treated A53T-L444P mice. Circles: A53T; squares: A53T-L444P. Open symbols: control; filled symbols: PQ + IR.</p>
Full article ">Figure 11
<p>(<b>A</b>) In the open field, activity levels were higher in A53T than A54T-L444P mice (<span class="html-italic">F</span>(1,81) = 4.091, * <span class="html-italic">p</span> = 0.046). (<b>B</b>) In the rotorod test, there was a genotype × treatment interaction (<span class="html-italic">F</span>(1,81) = 7.773, <span class="html-italic">p</span> = 0.007). DSS impaired rotorod performance in A53T-L444P mice (<span class="html-italic">F</span>(1,45) = 9.228, *** <span class="html-italic">p</span> = 0.004), but not in A53T mice. (<b>C</b>) DSS reduced spontaneous alternation in A53T-L444P mice (<span class="html-italic">F</span>(1,45) = 7.191, * <span class="html-italic">p</span> = 0.01). (<b>D</b>) A53T-L44P mice showed reduced activity levels in the Y-maze (<span class="html-italic">F</span>(1,81) = 5.336, * <span class="html-italic">p</span> = 0.023). (<b>E</b>) During the baseline period in the fear conditioning test, activity levels were lower in A53T-L44P than A53T mice (<span class="html-italic">F</span>(1,81) = 7.565, ** <span class="html-italic">p</span> = 0.007). (<b>F</b>) In males, A53T-L44P mice showed lower activity levels than A53T mice during the baseline period (<span class="html-italic">F</span>(1,41) = 6.727, * <span class="html-italic">p</span> = 0.013). (<b>G</b>) In females, there was a treatment × genotype interaction (<span class="html-italic">F</span>(1,40) = 4.168, <span class="html-italic">p</span> = 0.048). DSS increased activity level in A53T-L44P females (<span class="html-italic">F</span>(1,22) = 5.119, * <span class="html-italic">p</span> = 0.034). (<b>H</b>) For the freezing levels during the ISI, there was a genotype × treatment × sex interaction (<span class="html-italic">F</span>(1,81) = 4.661, <span class="html-italic">p</span> = 0.034). (<b>I</b>) A53T-L44P mice had lower activity levels than A53T mice during the ISI (<span class="html-italic">F</span>(1,81) = 4.564, * <span class="html-italic">p</span> = 0.036). (<b>J</b>) When activity levels during the tones were analyzed, there was a genotype × treatment × sex interaction (<span class="html-italic">F</span>(1,81) = 8.199, <span class="html-italic">p</span> = 0.005). (<b>K</b>) A53T-L44P mice had lower activity levels than A53T mice during the contextual fear memory test (<span class="html-italic">F</span>(1,85) = 4.677, * <span class="html-italic">p</span> = 0.033). Circles: A53T; squares: A53T-L444P. Open symbols: H<sub>2</sub>O; filled symbols: DSS.</p>
Full article ">Figure 12
<p>Capscale ordinations reveal genotypic and exposure dependent associations between the composition of the gut microbiome and behavior measures. (<b>A</b>) A constrained ordination illustrates the association between the taxonomic beta-diversity of the gut microbiome, as measured by the weighted unifrac metric, and PQ exposure, genotype, and the total distance moved during the third day of an open field test. (<b>B</b>) An analogous ordination illustrating the association between the taxonomic beta-diversity of the gut microbiome, as measured by the Bray–Curtis dissimilarity metric, and PQ exposure, genotype, and the total distance moved during the third day of an open field test. For both panels, arrows indicate the direction in ordination space that corresponds to the vector of association between beta-diversity and the behavioral covariate (i.e., arrowheads point towards samples with higher behavioral scores) for a given set of sample sets: control treatment and control genotype (solid grey lines), control treatment and L444P genotype (dashed grey lines), chemical exposure and control genotype (solid black lines), and chemical exposure and L444P genotype (dashed black lines). The different directionality of vectors indicates interaction effects of these various features on the association between microbiome composition and behavior, and these differences are supported by permanova tests (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
0 pages, 2937 KiB  
Article
Frameshift Variant in AMPD2 in Cirneco dell’Etna Dogs with Retinopathy and Tremors
by Leonardo Murgiano, Jessica K. Niggel, Leontine Benedicenti, Matteo Cortellari, Arianna Bionda, Paola Crepaldi, Luigi Liotta, Geoffrey K. Aguirre, William A. Beltran and Gustavo D. Aguirre
Genes 2024, 15(2), 238; https://doi.org/10.3390/genes15020238 - 13 Feb 2024
Viewed by 1473
Abstract
While the manifestations of many inherited retinal disorders are limited to loss of vision, others are part of a syndrome that affects multiple tissues, particularly the nervous system. Most syndromic retinal disorders are thought to be recessively inherited. Two dogs out of a [...] Read more.
While the manifestations of many inherited retinal disorders are limited to loss of vision, others are part of a syndrome that affects multiple tissues, particularly the nervous system. Most syndromic retinal disorders are thought to be recessively inherited. Two dogs out of a litter of Cirneco dell′ Etna dogs, both males, showed signs of retinal degeneration, along with tremors and signs described as either atypical seizures or paroxysmal dyskinesias, while the other two male littermates were normal. We named this oculo-neurological syndrome CONS (Cirneco oculo-neurological syndrome), and undertook homozygosity mapping and whole-genome sequencing to determine its potential genetic etiology. Notably, we detected a 1-bp deletion in chromosome 6 that was predicted to cause a frameshift and premature stop codon within the canine AMPD2 gene, which encodes adenosine monophosphate deaminase, an enzyme that converts adenosine 5′-monophosphate (AMP) to inosine 5’-monophosphate (IMP). Genotyping of the available Cirneco population suggested perfect segregation between cases and controls for the variant. Moreover, this variant was absent in canine genomic databases comprised of thousands of unaffected dogs. The AMPD2 genetic variant we identified in dogs presents with retinal manifestations, adding to the spectrum of neurological manifestations associated with AMPD2 variants in humans. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Retinal and cerebral phenotype. Left (<b>A</b>) and right (<b>B</b>) fundus pictures of a case (CRN4). Hyperreflectivity and marked attenuation/loss of retinal vessels along with pale atrophic optic discs indicate advanced stages of retinal degeneration. Sclerosis of the choroidal vessels is also present in the inferior non-tapetal region, accompanied by segmental choroid retinal atrophy. MRI of CRN4 (<b>C</b>) and CRN6 (<b>D</b>). (<b>C</b>) Transverse T2 FLAIR image at the level of the thalamus, where bilateral and symmetrical hyperintensities affecting the periventricular white matter tracts are depicted (arrows). (<b>D</b>): Transverse T2 weighted image at the level of the thalamus. In this case, no abnormalities were observed in the brain parenchyma. Moderate-to-severe left temporalis muscle atrophy is evident (arrow).</p>
Full article ">Figure 2
<p>Family tree and mapping. (<b>A</b>) Family tree of the cases. The four siblings shown include the two cases. Homozygous cases CRN4 and CRN6 are shown with a red-filled shape. Carriers (CRN1 and CRN5) and wild-type (CRN7) controls are shown with a half-filled and empty shape, respectively. Males are indicated with a square, females with a circle. Missing samples that we were unable to genotype are indicated with a diagonal bar. The carrier grandmother CRN1 is shared by both parents. (<b>B</b>) Mapping of the candidate regions. Homozygous regions exclusive for the cases are shown in red. The interval containing the candidate variant is indicated with an asterisk.</p>
Full article ">Figure 3
<p>Sequencing of the putative causative variant. (<b>A</b>) Whole genome sequencing of two affected Cirneco (CRN4 and CRN6) and of unrelated retinal RNA-seq (confirming the exon and transcript in retina) shown in Integrate Genome Viewer. Note the 1-bp deletion (g.42,698,170delC). (<b>B</b>) Sanger sequencing of a control (top) a carrier (middle) and a case (bottom) showing the deletion of the coding C. Variant indicated with a red arrow.</p>
Full article ">Figure 4
<p>Position of the proposed causative variant. (<b>A</b>) Position of the c.2,131delG variant in the 15th exon of the canine <span class="html-italic">AMPD2</span> transcript. Exons shown as re-annotated from canine retinal RNA-seq. (<b>B</b>) Human AMPD2 protein with the position of the AMPD2 mutations associated with neurological diseases in Homo sapiens as reported in the literature [<a href="#B52-genes-15-00238" class="html-bibr">52</a>,<a href="#B53-genes-15-00238" class="html-bibr">53</a>,<a href="#B54-genes-15-00238" class="html-bibr">54</a>,<a href="#B55-genes-15-00238" class="html-bibr">55</a>,<a href="#B56-genes-15-00238" class="html-bibr">56</a>,<a href="#B57-genes-15-00238" class="html-bibr">57</a>]. The canine AMPD2 variant (position as realigned to the human protein) is reported in red and with the larger arrow. As for most pathological human variants, the mutation we report occurs within the AMPD domain.</p>
Full article ">Figure 5
<p>(<b>A</b>) Representation of the first two principal components (PCs) of the multidimensional scaling analysis of the individual identity-by-state distances. (<b>B</b>) Best-fitting model (number of clusters K = 23) obtained from the admixture analysis. Each bar represents a subject and each color a different cluster.</p>
Full article ">
20 pages, 3212 KiB  
Article
Prolonged Antibiotic Use in a Preclinical Model of Gulf War Chronic Multisymptom-Illness Causes Renal Fibrosis-like Pathology via Increased micro-RNA 21-Induced PTEN Inhibition That Is Correlated with Low Host Lachnospiraceae Abundance
by Ayushi Trivedi, Dipro Bose, Punnag Saha, Subhajit Roy, Madhura More, Jonathan Skupsky, Nancy G. Klimas and Saurabh Chatterjee
Cells 2024, 13(1), 56; https://doi.org/10.3390/cells13010056 - 27 Dec 2023
Cited by 1 | Viewed by 1511
Abstract
Gulf War (GW) veterans show gastrointestinal disturbances and gut dysbiosis. Prolonged antibiotic treatments commonly employed in veterans, especially the use of fluoroquinolones and aminoglycosides, have also been associated with dysbiosis. This study investigates the effect of prolonged antibiotic exposure on risks of adverse [...] Read more.
Gulf War (GW) veterans show gastrointestinal disturbances and gut dysbiosis. Prolonged antibiotic treatments commonly employed in veterans, especially the use of fluoroquinolones and aminoglycosides, have also been associated with dysbiosis. This study investigates the effect of prolonged antibiotic exposure on risks of adverse renal pathology and its association with gut bacterial species abundance in underlying GWI and aims to uncover the molecular mechanisms leading to possible renal dysfunction with aging. Using a GWI mouse model, administration of a prolonged antibiotic regimen involving neomycin and enrofloxacin treatment for 5 months showed an exacerbated renal inflammation with increased NF-κB activation and pro-inflammatory cytokines levels. Involvement of the high mobility group 1 (HMGB1)-mediated receptor for advanced glycation end products (RAGE) activation triggered an inflammatory phenotype and increased transforming growth factor-β (TGF-β) production. Mechanistically, TGF-β- induced microRNA-21 upregulation in the renal tissue leads to decreased phosphatase and tensin homolog (PTEN) expression. The above event led to the activation of protein kinase-B (AKT) signaling, resulting in increased fibronectin production and fibrosis-like pathology. Importantly, the increased miR-21 was associated with low levels of Lachnospiraceae in the host gut which is also a key to heightened HMGB1-mediated inflammation. Overall, though correlative, the study highlights the complex interplay between GWI, host gut dysbiosis, prolonged antibiotics usage, and renal pathology via miR-21/PTEN/AKT signaling. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Analysis of Western blot data revealed that prolonged antibiotic administration (neomycin and enrofloxacin) in GWI mice resulted in increased kidney inflammation and elevated levels of pro-inflammatory cytokines, namely, IL-1β and IL-17A. Representative (<b>A</b>) depicts protein expression of phosphorylated-p65, total-p65, pro-IL-1β, cleaved IL-1 β, IL-17A, and β-actin in kidney tissue of CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). (<b>B</b>–<b>D</b>) represents densitometry analysis of phosphorylated-p65 normalized with total-p65 (<b>B</b>), cleaved IL-1β normalized with pro-IL-1β (<b>C</b>), and IL-17A normalized with β-actin (<b>D</b>) in CONTROL, GWI, GWI+AB, and AB mice plotted as bar graph. The data are presented as the mean ±  SD (SD: standard deviation) and statistical significance was measured using one-way ANOVA between all the groups, with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">Figure 2
<p>Immunohistochemical analysis and mRNA expression levels demonstrated that prolonged antibiotic administration (neomycin and enrofloxacin) in GWI mice led to an increase in the production of pro-inflammatory cytokines. Representative immunohistochemistry images (<b>A</b>) depict immunoreactivity of TNF-α in kidney sections from CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). Images were captured at a 40× magnification (scale bar = 50 µm). Immunoreactivity was denoted by the presence of red arrows and morphometric analysis was calculated as %ROI. (<b>B</b>) The bar graph represents the morphometric analysis of TNF-α in CONTROL, GWI, GWI+AB, and AB mice. The data are presented as the mean ±  SD (SD: standard deviation) of %ROI (mean value calculated from five different fields in each sample). Bar graphs (<b>C</b>–<b>F</b>) illustrate the normalized mRNA expression levels of different pro-inflammatory cytokines, namely IL-1 β (<b>C</b>), IL-17A (<b>D</b>), and TNF-α (<b>E</b>), and chemokines, specifically MCP-1 (<b>F</b>) against GAPDH. These expression levels are presented as fold changes relative to the CONTROL derived from kidney tissue samples of CONTROL, GWI, GWI+AB, and AB mice. The experiment was performed in triplicates and the data are represented as mean ± SEM (SEM: standard error mean). Statistical significance was determined using one-way ANOVA with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">Figure 3
<p>Immunofluorescence analysis demonstrated that prolonged antibiotic administration in GWI mice led to an augmentation in RAGE activation mediated by HMGB1. Representative (<b>A</b>) immunofluorescence images depict co-localization events (yellow) of HMGB1 (in red) and RAGE (in green) within kidney sections from CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). The kidney sections were additionally counterstained with DAPI (in blue), and all images were acquired at a 40× magnification (scale bar = 50 µm). Immunoreactivity was denoted by the presence of white arrows and morphometric analysis was calculated as %ROI. (<b>B</b>) The bar graph represents HMGB1-RAGE co-localization events in CONTROL, GWI, GWI+AB, and AB mice. The data are presented as the mean ±  SD (SD: standard deviation) of %ROI (mean value calculated from five different fields in each sample) and statistical significance was measured using one-way ANOVA between all the groups, with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">Figure 4
<p>Immunohistochemical analysis demonstrated that prolonged antibiotic administration in GWI mice led to an increase in renal TGF-β production. Representative immunohistochemistry images (<b>A</b>) depict immunoreactivity of TGF-β in kidney sections from CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). Images were captured at a 40× magnification (scale bar = 50 µm). Immunoreactivity was denoted by the presence of red arrows and morphometric analysis was calculated as %ROI. (<b>B</b>) The bar graph represents a morphometric analysis of TGF-β in CONTROL, GWI, GWI+AB, and AB mice. The data are presented as the mean ± SD (SD: standard deviation) of %ROI (mean value calculated from five different fields in each sample). Statistical significance was determined using one-way ANOVA with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">Figure 5
<p>Prolonged antibiotic administration in GWI mice mediated the upregulation of micro-RNA 21 (miR-21), a signature miRNA for renal fibrosis, and correlated with <span class="html-italic">Lachnospiraceae</span> abundance. (<b>A</b>) represents miR-21 expression analyzed by qRT-PCR in the kidney tissue of CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). The miR-21 expression was normalized using miR-103-3p as endogenous control. The bar graph is represented as fold change against CONTROL. (<b>B</b>) represents a bar graph showing the percent relative abundance of <span class="html-italic">Lachnospiraceae</span> spp. in CONTROL, GWI, GWI+AB, and -AB groups following whole genome sequencing of mouse fecal pellets. (<b>C</b>) depicts the correlation analysis of <span class="html-italic">Lachnospiraceae</span> spp. relative abundance and miR-21 levels in renal tissue of CONTROL, GWI, GWI+AB, and AB groups. Pearson’s linear regression is shown in red with 95% confidence bands. The experiment was performed in triplicates and the data are represented as mean ± SEM (SEM: standard error mean). Statistical significance was determined using one-way ANOVA with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">Figure 6
<p>The protein expression of PTEN, a target of miR-21, is perturbed due to prolonged antibiotic treatment in GWI mice, thereby modulating AKT signaling, as PTEN functions as a negative regulator of AKT signaling. (<b>A</b>) represents Western blot analysis of the relative protein expression of PTEN, phosphorylated-AKT, total-AKT, and β-actin in kidney tissue of CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). (<b>B</b>,<b>C</b>) represents a bar graph illustrating the densitometry analysis of PTEN normalized with β-actin (<b>B</b>) and phosphorylated-AKT normalized with total-AKT (<b>C</b>) in CONTROL, GWI, GWI+AB, and AB mice plotted as a bar graph. The data are presented as the mean ± SD (SD: standard deviation) and statistical significance was measured using one-way ANOVA between all the groups, with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">Figure 7
<p>Prolonged antibiotic treatment in GWI mice triggers the activation of AKT signaling, resulting in increased extracellular matrix deposition (ECM) and the induction of epithelial–mesenchymal transition (EMT)-like phenotype. Representative immunohistochemistry images (<b>A</b>) depict immunoreactivity of Fibronectin and α-SMA in kidney sections from CONTROL, GWI (persistent Gulf War Illness), GWI+AB (persistent Gulf War Illness with prolonged antibiotics exposure), and AB mice (prolonged antibiotics exposure only) (n = 6/group). Images were captured at a 40× magnification (scale bar = 50 µm). Immunoreactivity was denoted by the presence of red arrows and morphometric analysis was calculated as %ROI. (<b>B</b>,<b>C</b>) represents a bar graph illustrating the morphometric analysis of Fibronectin (<a href="#cells-13-00056-f006" class="html-fig">Figure 6</a>B) and α-SMA (Figure. 6C) in CONTROL, GWI, GWI+AB, and AB mice. The data are presented as the mean ±  SD (SD: standard deviation) of %ROI (mean value calculated from five different fields in each sample). Statistical significance was determined using one-way ANOVA with the Bonferroni–Dunn post hoc test. <span class="html-italic">p</span> &lt; 0.05 was considered statistically significant.</p>
Full article ">
21 pages, 3913 KiB  
Article
Severely Damaged Freeze-Injured Skeletal Muscle Reveals Functional Impairment, Inadequate Repair, and Opportunity for Human Stem Cell Application
by Daniela Fioretti, Mario Ledda, Sandra Iurescia, Raffaella Carletti, Cira Di Gioia, Maria Grazia Lolli, Rodolfo Marchese, Antonella Lisi and Monica Rinaldi
Biomedicines 2024, 12(1), 30; https://doi.org/10.3390/biomedicines12010030 - 21 Dec 2023
Viewed by 1053
Abstract
Background: The regeneration of severe traumatic muscle injuries is an unsolved medical need that is relevant for civilian and military medicine. In this work, we produced a critically sized nonhealing muscle defect in a mouse model to investigate muscle degeneration/healing phases. Materials and [...] Read more.
Background: The regeneration of severe traumatic muscle injuries is an unsolved medical need that is relevant for civilian and military medicine. In this work, we produced a critically sized nonhealing muscle defect in a mouse model to investigate muscle degeneration/healing phases. Materials and methods: We caused a freeze injury (FI) in the biceps femoris of C57BL/6N mice. From day 1 to day 25 post-injury, we conducted histological/morphometric examinations, an analysis of the expression of genes involved in inflammation/regeneration, and an in vivo functional evaluation. Results: We found that FI activates cytosolic DNA sensing and inflammatory responses. Persistent macrophage infiltration, the prolonged expression of eMHC, the presence of centrally nucleated myofibers, and the presence of PAX7+ satellite cells at late time points and with chronic physical impairment indicated inadequate repair. By looking at stem-cell-based therapeutic protocols of muscle repair, we investigated the crosstalk between M1-biased macrophages and human amniotic mesenchymal stem cells (hAMSCs) in vitro. We demonstrated their reciprocal paracrine effects where hAMSCs induced a shift of M1 macrophages into an anti-inflammatory phenotype, and M1 macrophages promoted an increase in the expression of hAMSC immunomodulatory factors. Conclusions: Our findings support the rationale for the future use of our injury model to exploit the full potential of in vivo hAMSC transplantation following severe traumatic injuries. Full article
(This article belongs to the Topic Animal Models of Human Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>FI protocol produced significant deficits in the animal’s functional ability. (<b>A</b>) Latency to fall in rotarod test at 1, 2, 4, 7, and 25 days after injury. The average period (±SD) of each group (4 mice) for the 3 repeated tests is shown. Uninjured mice represent reference control with 100% functionality. (<b>B</b>) The average number of paw slips (±SD) for 3 repeated tests is represented by a histogram chart. Uninjured shows the minimum number of slips by the healthy group. One-way ANOVA with a post hoc test. <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 vs. uninjured.</p>
Full article ">Figure 2
<p>FI provoked histological and morphometric modifications in mouse <span class="html-italic">biceps femoris</span> muscle tissue. (<b>A</b>) Representative H&amp;E and Sirius Red staining (for collagen deposits) of injured muscle tissue sections at each sampling time point after FI. Both uninjured tissues and areas peripheral to injury were examined. Scale bars: 100 µm (20×), 200 µm (40×). Arrows show the damage boundary line. (<b>B</b>) Injury size assessment at each sampling time point after FI (data are expressed as the percentage ratio of damaged area vs. total area); the dotted line indicates the critical threshold point (15%) of the muscle defect as described by Anderson et al. [<a href="#B3-biomedicines-12-00030" class="html-bibr">3</a>]. (<b>C</b>) Collagen (connective tissue) quantification in the muscle tissue sections at each time point after FI. Graph bars represent the percentage of the injured area occupied by connective tissue. Data are expressed as mean ± SD. ANOVA with a post hoc test; <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 vs. uninjured and # <span class="html-italic">p</span> &lt; 0.05 vs. day 4.</p>
Full article ">Figure 3
<p>Muscle regeneration parameter assessment after FI. Histograms representing quantification of cross-sectional area (CSA) of myofibers, expressed in µm<sup>2</sup>; frequency distribution of myofiber CSA. (<b>A</b>,<b>B</b>) Freeze-injured site, (<b>D</b>,<b>E</b>) region surrounding the lesion, (<b>C</b>) quantitative evaluation of muscle fibers in transverse section with centrally positioned nuclei in the regenerating area. (<b>F</b>) IHC identification of PAX7<sup>+</sup> satellite cells (brown reaction) at each time point after FI. In the injured samples, images were acquired at the periphery of the lesioned area. The scale bar represents 100 µm. Relative histograms representing the percentage of PAX7<sup>+</sup> cells were generated from the same data. n = 4 mice in each group. Data are expressed as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 vs. uninjured.</p>
Full article ">Figure 4
<p>FI activates IRF3-dependent cytosolic DNA sensing and inflammatory response. Expression levels of Ifnβ1, Cxcl10, Ifit1, Ifit2, Ifit3, Oasl2, Irf7, Rsda2, Tmem173. Uninjured muscles were used as controls. One-way ANOVA with a post hoc test. Data are expressed as mean ± SD. <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 vs. uninjured and # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.001 vs. day 1 and - <span class="html-italic">p</span> &lt; 0.05 vs. day 4.</p>
Full article ">Figure 5
<p>FI induced substantial inflammatory cell infiltration in damaged muscle tissue areas. (<b>A</b>) Quantitative evaluation of neutrophils after FI compared with uninjured control. Data are expressed as mean ± SD. (<b>B</b>) Quantitative evaluation of CD68<sup>+</sup> macrophages. Data are expressed as mean ± SD. (<b>C</b>) CD68<sup>+</sup> immunohistochemical staining of macrophage cells (brown reaction). Representative muscle sections in uninjured and injured tissue areas at early (day 1), middle (day 4), and late (day 25) stages after FI; (<b>D</b>) CD163<sup>+</sup> and CD206<sup>+</sup> immunohistochemical staining of macrophage cells (brown reaction). Representative muscle sections in injured tissue area at 4 and 25 days after FI, respectively (40×, scale bar 100 µm). Data are expressed as mean ± SD. <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 vs. uninjured and # <span class="html-italic">p</span> &lt; 0.05 vs. day 2.</p>
Full article ">Figure 6
<p>FI-induced expression of pro-inflammatory and anti-inflammatory markers in damaged muscle tissue. Expression levels of pro-inflammatory (Tnf-α, Il-6, Ccl2) and anti-inflammatory (Il-10, Arg1, and CD206) markers. Uninjured mice were used as controls. Statistical significance was evaluated by one-way ANOVA with a post hoc test. <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 vs. uninjured.</p>
Full article ">Figure 7
<p>FI-induced expression of muscle regeneration markers in damaged mice. Expression levels of myogenic regeneration markers, eMHC, Pax7, Myod, and Myog. Uninjured mice were used as controls. Statistical significance was evaluated by one-way ANOVA with a post hoc test. Data are expressed as mean ± SD. <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 vs. uninjured.</p>
Full article ">Figure 8
<p>(<b>A</b>) hAMSCs affected mRNA expression of pro-inflammatory and anti-inflammatory markers in M1-polarized mouse RAW 264.7 macrophages. mRNA expression levels of pro-inflammatory and anti-inflammatory markers (Nos2, Tnf-α, Il-10) in naïve (naïve Mφ) and M1-polarized macrophages RAW 264.7 (M1 Mφ). Statistical significance was evaluated by Student’s <span class="html-italic">t</span>-test. * vs. M1 Mφ. (<b>B</b>) M1-polarized (M1 Mφ) RAW 264.7 macrophages were able to trigger immunomodulatory molecule transcription in hAMSCs. mRNA expression levels of immunomodulatory markers (IDO, COX-2, and HGF) in primed hAMSCs compared with hAMSCs alone. Statistical significance was evaluated by one-way ANOVA with a post hoc test. Data are expressed as mean ± SD. <span class="html-italic">n</span> = 4 mice in each group. * <span class="html-italic">p</span> &lt; 0.05 vs. hAMSCs.</p>
Full article ">
Back to TopTop