[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,270)

Search Parameters:
Keywords = nerve injury

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 10018 KiB  
Article
Variations and Asymmetry in Sacral Ventral Rami Contributions to the Bladder
by Rebeccah R. Overton, Istvan P. Tamas, Emily P. Day, Nagat Frara, Michel A. Pontari, Susan B. Fecho, Steven N. Popoff and Mary F. Barbe
Diagnostics 2025, 15(1), 102; https://doi.org/10.3390/diagnostics15010102 - 3 Jan 2025
Viewed by 293
Abstract
Background/Objectives: We have demonstrated in human cadavers and canines that nerve transfer to bladder vesical nerve branches is technically feasible for bladder reinnervation after nerve injury. We further clarify here that sacral (S) ventral rami contribute to these vesical branches in 36 pelvic [...] Read more.
Background/Objectives: We have demonstrated in human cadavers and canines that nerve transfer to bladder vesical nerve branches is technically feasible for bladder reinnervation after nerve injury. We further clarify here that sacral (S) ventral rami contribute to these vesical branches in 36 pelvic sides (in 22 human cadavers). Methods: Gross post-mortem visualization and open anterior abdominal approaches were used, as was micro-CT of sacral nerve bundles, for further confirmation when needed. Results: Considerable between and within-subject variation was observed. Sacral (S) ventral rami contributions to vesical nerves were observed as shared contributions from several rami or, in a few cases, from single rami: S2 alone (6%), S3 alone (6%), S2 and S3 (28%), S3 and S4 (28%), S2–S4, 14%, L5 in combination with S1–S4 (6%), S1 and S2 (6%), and S3–S5 (3%). The most common contributor to these shared or single rami contributions was from the S3 ventral ramus, which contributed 100% of the time on the left side and 79% on the right side. Side-to-side asymmetry was observed in 10 of 14 cadavers examined bilaterally (71%). Conclusions: This characterization of the anatomical variation in sacral ventral rami contributions to the bladder will ultimately aid in developing therapeutics for patients with bladder dysfunction. Full article
(This article belongs to the Special Issue Advances in Anatomy—Third Edition)
Show Figures

Figure 1

Figure 1
<p>Percent individual or exclusive/shared contributions of lumbar and sacral (L5–S5) ventral rami contributions to the bladder. (<b>A</b>) Segmental contributions from each individual segment, with right and left side data combined. (<b>B</b>) Root contributions to the bladder (exclusive or shared), with right and left side data combined. (<b>C</b>) Segmental Contributions from each individual segment, separated by left vs. right pelvic plexus. (<b>D</b>) Root contributions to the bladder (exclusive or shared), separated by left vs. right pelvic plexus. “0” denotes no contribution. Most of the input was from S2, S3, and S4. Raw data is shown in <a href="#diagnostics-15-00102-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 2
<p>Example of S3 ventral ramus contributions to the bladder. (<b>A</b>) A hemisected pelvis of a fixed cadaver, shown looking from the midline (vertebrae) laterally (ureter is marked by a red vessel loop). (<b>B</b>) Potential sacral ventral rami contributions are labeled with a yellow tissue dye (S2, S3 and S4 are labeled, although only S2 and S3 appeared to be contributors at this point in the dissection). Their contributions to the vesical branches are grossly marked with a dark red tissue dye for later visualization after removal as a tissue block. (<b>C</b>) Tissue block with S2 and S3 ventral rami and vesical branches with adjacent fat, blood vessels and connective tissues. (<b>D</b>) Micro-CT was used to scan the entire Lugol-stained tissue block. Longitudinally oriented 3D image shown. (<b>E</b>,<b>F</b>) Cross-sectional images from the vesical nerve ends of the tissue block. Vesical branches are indicated in both panels with arrows; vesical branches are highlighted with pink in panel (<b>E</b>), and are indicated with arrows in both panels (<b>E</b>,<b>F</b>). (<b>G</b>) Close-up of the ventral rami at the proximal spinal cord end from another image. Both S2 and S3 ventral roots are shown. As shown in panel (<b>E</b>), the S2 root could not be traced to the vesical branches.</p>
Full article ">Figure 3
<p>Examples of S1 and S2, and S2 and S3, ventral rami contributions to the bladder in two different cadavers. (<b>A</b>) The hemisected pelvis of a fixed cadaver in which ventral rami from both S1 and S2 contributed branches to the bladder (a red wire surrounds these roots). Scissor tips show nerves passing to the vesical branches of the pelvic nerve (overlying the green wire and indicated with an arrow). The ureter is delineated by a yellow wire. (<b>B</b>) A diagram of panel (<b>A</b>) in which the sacral contributions are colored yellow and the bladder as pink. (<b>C</b>) An intact pelvic region of an unfixed cadaver. The image shows a superior view of a fresh pelvic region opened using an anterior abdominal approach. The outer wall of the bladder is labeled; the ureter is indicated by a blue vessel loop. Vesical branches could be seen arising from both S2 and S3 ventral rami (arrows).</p>
Full article ">Figure 4
<p>Examples of S3 and S4 ventral rami contributions to the bladder in two different cadavers. (<b>A</b>) The hemisected pelvis of a fixed cadaver in which S3 and S4 ventral rami contribute branches to the bladder (encircled by a white wire). S3 had an extra slip arising adjacent to the main root. The ureter, located deep to this view, is encircled with a blue wire. The vesical branches were adjacent to the ureter in this cadaver. (<b>B</b>) Diagram of panel A in which the S3 and S4 ventral rami contributions to the bladder are colored yellow, and the bladder is colored pink.</p>
Full article ">Figure 5
<p>Images of multiple contributions from L5–S4 are more difficult to present as the L5 and S4 branches were quite small in circumference. Example of S2, S3 and S4 combined root contributions to the vesical branches of the bladder (indicated by path of arrows). (<b>A</b>) Hemisected right side pelvis of a fixed cadaver depicting S2–S4 ventral root contributions to the bladder, highlighted in yellow. The other sacral roots are indicated in pink, and the obturator nerve is highlighted in red. The forceps were used to elevate the bladder for this view. (<b>B</b>) The same image as in Panel (<b>A</b>) is shown unlabeled.</p>
Full article ">
14 pages, 4952 KiB  
Article
Efficacy of Photobiomodulation Therapy Utilizing 808 nm and 660 nm Alone and in Combination for Treatment of Paresthesia in Rats
by Ehsan Hajesmaelzade, Mohammad Mohammadi, Sina Kakooei, Luca Solimei, Stefano Benedicenti and Nasim Chiniforush
Biomedicines 2025, 13(1), 65; https://doi.org/10.3390/biomedicines13010065 - 30 Dec 2024
Viewed by 299
Abstract
Background/Objectives: This study assessed the efficacy of photobiomodulation therapy (PBM) by 808 nm and 660 nm alone and in combination for the treatment of paresthesia in rats. Methods: This animal study was conducted on 36 adult male Wistar rats. After general [...] Read more.
Background/Objectives: This study assessed the efficacy of photobiomodulation therapy (PBM) by 808 nm and 660 nm alone and in combination for the treatment of paresthesia in rats. Methods: This animal study was conducted on 36 adult male Wistar rats. After general anesthesia, the facial nerve of the right side of the face of rats was surgically exposed and pinched, returned in place, and sutured. The rats were randomly assigned to six groups (n = 6) of (I) no-intervention (control), (II) no-laser, (III) 808 nm laser (250 mW, 4 W/cm2, 20 s, 8 J/cm2, (IV) 660 nm laser (150 mW, 0.25 W/cm2, 32 s, 8 J/cm2, (V) 808 nm plus 660 nm laser with the original settings, and (VI) 808 nm plus 660 nm laser with half of the time and energy density. After 16 days, a biopsy sample was taken from the nerve injury site and underwent histological, histometric, and immunohistochemical assessments. Results: Significantly lower edema and congestion were seen in the combined laser group with original settings (p < 0.05); this group had no significant difference with the control group regarding degenerative changes of the nerve fibers and Schwann cells (p > 0.05). The 660 nm, and combined laser groups, had a significantly lower accumulation of inflammatory cells (p < 0.05). The number of blood vessels in combined laser groups was significantly lower than that in the no-laser group (p < 0.05). Conclusions: The results showed the positive efficacy of PBM by 808 nm and 660 nm lasers in resolution of inflammation and reduction of degenerative changes of Schwann cells and nerve fibers. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

Figure 1
<p>Histological micrographs of the control group; (<b>A</b>–<b>C</b>) H and E staining with ×10, ×20, and ×40 magnification, respectively; (<b>D</b>–<b>F</b>) S100 staining with ×10, ×20, and ×40 magnification, respectively.</p>
Full article ">Figure 2
<p>Histological micrographs of the no-laser group; (<b>A</b>–<b>C</b>) H and E staining with ×10, ×20, and ×40 magnification, respectively; (<b>D</b>–<b>F</b>) S100 staining with ×10, ×20, and ×40 magnification, respectively.</p>
Full article ">Figure 3
<p>Histological micrographs of 808 nm laser group with 20 s irradiation time and 8 J/cm<sup>2</sup> energy density; (<b>A</b>–<b>C</b>) H and E staining with ×10, ×20, and ×40 magnification, respectively; (<b>D</b>–<b>F</b>) S100 staining with ×10, ×20, and ×40 magnification, respectively.</p>
Full article ">Figure 4
<p>Histological micrographs of the 660 nm laser group with 20 s irradiation time and 8 J/cm<sup>2</sup> energy density; (<b>A</b>–<b>C</b>) H and E staining with ×10, ×20, and ×40 magnification, respectively; (<b>D</b>–<b>F</b>) S100 staining with ×10, ×20, and ×40 magnification, respectively.</p>
Full article ">Figure 5
<p>Histological micrographs of 808 nm and 660 nm combined laser group with half the time (10 s) and energy density (4 J/cm<sup>2</sup>); (<b>A</b>–<b>C</b>) H and E staining with ×10, ×20, and ×40 magnification, respectively; (<b>D</b>–<b>F</b>) S100 staining with ×10, ×20, and ×40 magnification, respectively.</p>
Full article ">Figure 6
<p>Histological micrographs of 808 nm and 660 nm combined laser group with original time (20 s) and energy density (8 J/cm<sup>2</sup>); (<b>A</b>–<b>C</b>) H and E staining with ×10, ×20, and ×40 magnification, respectively; (<b>D</b>–<b>F</b>) S100 staining with ×10, ×20, and ×40 magnification, respectively.</p>
Full article ">
22 pages, 13437 KiB  
Article
The Intrinsic Neuronal Activation of the CXCR4 Signaling Axis Is Associated with a Pro-Regenerative State in Cervical Primary Sensory Neurons Conditioned by a Sciatic Nerve Lesion
by Petr Dubový, Ivana Hradilová-Svíženská, Václav Brázda, Anna Jambrichová, Viktorie Svobodová and Marek Joukal
Int. J. Mol. Sci. 2025, 26(1), 193; https://doi.org/10.3390/ijms26010193 - 29 Dec 2024
Viewed by 342
Abstract
CXCL12 and CXCR4 proteins and mRNAs were monitored in the dorsal root ganglia (DRGs) of lumbar (L4–L5) and cervical (C7–C8) spinal segments of naïve rats, rats subjected to sham operation, and those undergoing unilateral complete sciatic nerve transection (CSNT) on post-operation day 7 [...] Read more.
CXCL12 and CXCR4 proteins and mRNAs were monitored in the dorsal root ganglia (DRGs) of lumbar (L4–L5) and cervical (C7–C8) spinal segments of naïve rats, rats subjected to sham operation, and those undergoing unilateral complete sciatic nerve transection (CSNT) on post-operation day 7 (POD7). Immunohistochemical, Western blot, and RT-PCR analyses revealed bilaterally increased levels of CXCR4 protein and mRNA in both lumbar and cervical DRG neurons after CSNT. Similarly, CXCL12 protein levels increased, and CXCL12 mRNA was upregulated primarily in lumbar DRGs ipsilateral to the nerve lesion. Intrathecal application of the CXCR4 inhibitor AMD3100 following CSNT reduced CXCL12 and CXCR4 protein levels in cervical DRG neurons, as well as the length of afferent axons regenerated distal to the ulnar nerve crush. Furthermore, treatment with the CXCR4 inhibitor decreased levels of activated Signal Transducer and Activator of Transcription 3 (STAT3), a critical transforming factor in the neuronal regeneration program. Administration of IL-6 increased CXCR4 levels, whereas the JAK2-dependent STAT3 phosphorylation inhibitor (AG490) conversely decreased CXCR4 levels. This indicates a link between the CXCL12/CXCR4 signaling axis and IL-6-induced activation of STAT3 in the sciatic nerve injury-induced pro-regenerative state of cervical DRG neurons. The role of CXCR4 signaling in the axon-promoting state of DRG neurons was confirmed through in vitro cultivation of primary sensory neurons in a medium supplemented with CXCL12, with or without AMD3100. The potential involvement of conditioned cervical DRG neurons in the induction of neuropathic pain is discussed. Full article
(This article belongs to the Special Issue Advances in Peripheral Nerve Regeneration)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Representative sections of DRGs from naïve rats (Naive) and rats undergoing sterile sham (Sham) and CSNT operations on POD7 (CSNT7D). DRGs from CSNT-operated rats were taken from lumbar (L4-DRG) and cervical (C7-DRG) segments on both ipsilateral (i) and contralateral (c) sides. All sections were immunostained for CXCL12 detection under identical conditions. Increased intensities of CXCL12 immunofluorescence were observed in the neuronal bodies (arrows) and satellite glial cells (arrowheads) of both lumbar and cervical DRGs compared with those from naïve or sham-operated rats. Scale bars = 40 µm. (<b>b</b>) The results of CXCL12-IF intensities measured in the neuronal cytoplasm of large (L), medium (M), and small (S) cervical DRG neurons from naïve rats (Naïve), as well as both ipsilateral and contralateral DRGs of Sham- and CSNT-operated rats for POD7. *** Significant differences (<span class="html-italic">p</span> &lt; 0.001) compared with sham-operated rats, as was determined using a Mann–Whitney U-test.</p>
Full article ">Figure 2
<p>(<b>a</b>) Representative sections of DRGs from naïve rat (Naive) and rats after sterile sham (Sham) and CSNT operations on POD7 (CSNT7D). DRGs from CSNT-operated rats were taken from lumbar (L4-DRG) and cervical (C7-DRG) segments on both ipsilateral (i) and contralateral (c) sides. All sections were immunostained for CXCR4 detection under identical conditions. Sciatic nerve lesions induced an increased intensity of CXCR4 immunofluorescence in neuronal nuclei (arrowheads) and a diffuse pattern in the bodies of some DRG neurons (arrows). The heightened intensities of CXCR4 immunofluorescence predominantly loaded nuclei of both DRG neurons and their satellite glial cells. Scale bars = 40 µm. (<b>b</b>) The results of CXCR4-IF intensities measured in the neuronal nuclei of the cervical (C) and lumbar (L) DRGs from both ipsilateral (i) and contralateral (c) sides removed from naïve rats (Naïve) as well as from Sham- and CSNT-operated rats on POD7. *** Significant differences (<span class="html-italic">p</span> &lt; 0.001) compared with sham-operated rats, as was determined using a Mann–Whitney U-test.</p>
Full article ">Figure 3
<p>Results of real-time PCR (RT-PCR) of relative CXCL12 and CXCR4 mRNA levels in DRGs of lumbar (L) and the cervical (C) spinal segments (L4–L5) and (C7–C8), respectively, from the ipsilateral (i) and contralateral (c) sides. Tissue samples were collected from naïve rats as well as from sham- and CSNT-operated rats at POD7 (n = 9 for each group). Relative expressions were calculated using Actin as the housekeeping gene and normalized to naïve controls. * Significant differences (<span class="html-italic">p</span> &lt; 0.05) compared with sham-operated rats were determined using a Mann–Whitney U-test.</p>
Full article ">Figure 4
<p>(<b>a</b>) The representative sections through the cervical (C7-DRG) and lumbar (L4-DRG) DRGs harvested from the ipsilateral (i) side of rats subjected to CSNT on POD7, and after intrathecal application of artificial cerebrospinal fluid (ACSF) or AMD3100. Sections were incubated under the same conditions to demonstrate CXCR4 and CXCL12 immunofluorescence. The results showed reduced intensities of both CXCL12 and CXCR4 immunofluorescence in DRG sections from rats treated with AMD3100 compared with those treated with ACSF. Changes in CXCR4 immunofluorescence intensities were observed in neuronal nuclei (arrowheads), while changes in CXCL12 immunofluorescence intensities were noted in the neuronal bodies (arrows) and satellite glial cells (arrowheads). Scale bars = 50 µm. (<b>b</b>) Representative blots of CXCR4 and CXCL12 proteins with equal protein loading confirmed by actin levels (Actin). Samples of DRGs from both lumbar (L) and cervical (C) segments were collected from naïve rats and from the ipsilateral (i) and contralateral (c) sides of rats undergoing CSNT on POD7, followed by intrathecal application of either artificial cerebrospinal fluid (ACSF) or AMD3100. (<b>c</b>) The densitometry results of CXCR4 and CXCL12 protein bands, normalized to Actin levels. Densities of CXCR4 and CXCL12 bands from the naïve DRGs were set as 1 for reference. *, ** denote significant differences (<span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01, respectively) compared with naïve controls, while †† indicates a significant difference (<span class="html-italic">p</span> &lt; 0.01) compared with ACSF-treated counterparts, as determined by the Mann–Whitney U-test.</p>
Full article ">Figure 5
<p>In vivo assay of the pro-regenerative state of cervical DRG neurons conditioned by unilateral CSNT on POD7. The results revealed that intrathecal application of AMD3100 significantly reduced the lengths of SCG10-immunopositive sensory axons regenerated distal to the crush point compared with rats subjected to CSNT and treated with ACSF. (<b>a</b>) Representative longitudinal sections through the ulnar nerve distal to the crush site following prior CSNT and intrathecal application of either ACSF or AMD3100. Arrows indicate the maximal length of regenerated axons. Scale bars = 250 µm. (<b>b</b>) The lengths of SCG10-immunopositive axons measured distal to the ulnar nerve crush (n = 4 for each group). ** indicates a significant difference (<span class="html-italic">p</span> &lt; 0.01) compared with rats treated with ACSF, as determined by the Mann–Whitney U-test.</p>
Full article ">Figure 6
<p>The primary sensory neurons isolated from cervical DRGs were cultivated in vitro (<b>a</b>) in control medium (Control), in medium with the addition of CXCL12 (CXCL12), and in medium supplemented with both CXCL12 and AMD3100 (CXCL12+AMD3100). Scale bars = 75 µm. (<b>b</b>) The measurement results showed that the addition of CXCL12 into the medium significantly increased the number and length of neurites per neuron. Conversely, the medium supplemented with CXCL12 and AMD3100 reduced the outgrowth of neurites. * denotes a significant difference (<span class="html-italic">p</span> &lt; 0.05) compared with DRG neurons cultivated in control medium; † indicates a significant difference (<span class="html-italic">p</span> &lt; 0.05) between DRG neurons treated with CXCL12 and CXCL12 + AMD3100 media.</p>
Full article ">Figure 7
<p>(<b>a</b>) The results of STAT3(Y705) immunofluorescence staining are present in representative sections of DRGs. Sections through the cervical (C7-DRG) harvested from a naïve rat (Naïve) and a rat subjected to CSNT on POD7 (CSNT7D). In addition, sections of the cervical (C7-DRGi) and lumbar (L4-DRGi) ganglia ipsilateral (i) to CSNT on POD7 were obtained from rats after intrathecal application of artificial cerebrospinal fluid (ACSF) or AMD3100. The sections were immunostained under identical conditions to detect STAT3(Y705) in the neuronal nuclei (arrowheads). Scale bars = 25 µm. (<b>b</b>) Intensities of STAT3(Y705) immunofluorescence measured in the nuclei of DRG neurons (n = 3 for each group) were significantly decreased in DRGs of L4–L5 and C7–C8 segments (L4–L5 DRG, C7–C8 DRG) from both ipsilateral (i) and contralateral (c) sides of rats undergoing CSNT followed intrathecal application of AMD3100, compared with those from ACSF-treated controls. **, *** indicate significant differences (<span class="html-italic">p</span> &lt; 0.01 or <span class="html-italic">p</span> &lt; 0.001, respectively) compared with DRGs of ACSF-treated rats as determined by the Mann–Whitney U-test.</p>
Full article ">Figure 8
<p>(<b>a</b>) The representative sections of cervical DRGs (C7) harvested from rats subjected to CSNT on POD7 followed by intrathecal application of ACSF, IL-6 or JAK2 inhibitor AG490. Sections were immunostained for CXCR4 under identical conditions. Scale bars = 25 µm. (<b>b</b>) Measurement of immunofluorescence intensities in neuronal nuclei demonstrated that intrathecal administration of IL-6 increased, while JAK2 inhibitor significantly decreased, the intensities of CXCR4-IF compared to those of control rats treated with ACSF (n = 3 for each group). ***, * denote significant differences (<span class="html-italic">p</span> &lt; 0.001 or <span class="html-italic">p</span> &lt; 0.05, respectively) compared with DRGs of ACSF-treated rats, as determined by the Mann–Whitney U-test.</p>
Full article ">Figure 9
<p>The representative longitudinal sections distal to the UN crush from rats with a prior sciatic nerve lesion for 7 days. The sections were double-immunostained for CXCR4 and GAP43 (<b>a</b>) as well as CXCL12 and GAP43 (<b>b</b>) or GFAP (<b>c</b>). Merged pictures detected immunopositivity for CXCR4 in the growth cones (long arrows) as well as CXCL12 in non-neuronal cells that displayed GFAP immunopositivity indicating their Schwann cell origin (arrowheads). Scale bars = 30 µm.</p>
Full article ">Figure 10
<p>A diagram illustrating the rat experimental groups, timelines, and samples collected from cervical (C7–C8) and lumbar (L4–L5) DRGs, as well as the segments distal to UN crush, for analyses. (<b>A</b>) Samples were used for immunohistochemical analysis of CXCL12 and CXCR4 proteins and their mRNAs using RT-PCR. (<b>B</b>) Samples were analyzed by immunohistochemistry and western blot for CXCL12 and CXCR4 proteins, as well as immunohistochemical analysis of STAT3. (<b>C</b>) Nerve segments distal to the UN crush were used for SCG10 immunohistochemical analysis. (<b>D</b>) Cervical DRGs were analyzed for CXCR4 in neuronal nuclei by immunohistochemistry.</p>
Full article ">
15 pages, 2776 KiB  
Review
Preoperative Vascular and Cranial Nerve Imaging in Skull Base Tumors
by Akinari Yamano, Masahide Matsuda and Eiichi Ishikawa
Cancers 2025, 17(1), 62; https://doi.org/10.3390/cancers17010062 - 28 Dec 2024
Viewed by 409
Abstract
Skull base tumors such as meningiomas and schwannomas are often pathologically benign. However, surgery for these tumors poses significant challenges because of their proximity to critical structures such as the brainstem, cerebral arteries, veins, and cranial nerves. These structures are compressed or encased [...] Read more.
Skull base tumors such as meningiomas and schwannomas are often pathologically benign. However, surgery for these tumors poses significant challenges because of their proximity to critical structures such as the brainstem, cerebral arteries, veins, and cranial nerves. These structures are compressed or encased by the tumor as they grow, increasing the risk of unintended injury to these structures, which can potentially lead to severe neurological deficits. Preoperative imaging is crucial for assessing the tumor size, location, and its relationship with adjacent vital structures. This study reviews advanced imaging techniques that allow detailed visualization of vascular structures and cranial nerves. Contrast-enhanced computed tomography and digital subtraction angiography are optimal for evaluating vascular structures, whereas magnetic resonance imaging (MRI) with high-resolution T2-weighted images and diffusion tensor imaging are optimal for evaluating cranial nerves. These methods help surgeons plan tumor resection strategies, including surgical approaches, more precisely. An accurate preoperative assessment can contribute to safe tumor resection and preserve neurological function. Additionally, we report the MRI contrast defect sign in skull base meningiomas, which suggests cranial nerve penetration through the tumor. This is an essential finding for inferring the course of cranial nerves completely encased within the tumor. These preoperative imaging techniques have the potential to improve the outcomes of patients with skull base tumors. Furthermore, this study highlights the importance of multimodal imaging approaches and discusses future directions for imaging technology that could further develop preoperative surgical simulations and improve the quality of complex skull base tumor surgeries. Full article
(This article belongs to the Special Issue Advances in Tumor Vascular Imaging)
Show Figures

Figure 1

Figure 1
<p>Modality selection and its advantages and disadvantages in preoperative vascular and cranial nerve imaging.</p>
Full article ">Figure 2
<p>Illustrative cases of skull base meningiomas with cranial nerve penetration. (<b>a</b>) T1-weighted image with contrast of a patient with a left petroclival meningioma. The yellow arrowhead shows the contrast defect in the abducens nerve. (<b>b</b>) Intraoperative view of the abducens nerve penetrating the tumor (yellow arrow). (<b>c</b>) T1-weighted image showing the contrast of the patient with a right petrous meningioma. The yellow arrowhead indicates a contrast defect in the lower cranial nerve. (<b>d</b>) Intraoperative view of the glossopharyngeal nerve penetrating through the tumor (yellow arrow).</p>
Full article ">
21 pages, 273 KiB  
Article
Prospective Voice Assessment After Thyroidectomy Without Recurrent Laryngeal Nerve Injury
by Ivana Šimić Prgomet, Stjepan Frkanec, Ika Gugić Radojković and Drago Prgomet
Diagnostics 2025, 15(1), 37; https://doi.org/10.3390/diagnostics15010037 - 27 Dec 2024
Viewed by 282
Abstract
Background: Thyroidectomy, a surgical procedure for thyroid disorders, is associated with postoperative voice changes, even in cases without recurrent laryngeal nerve (RLN) injury. Our study evaluates the prevalence and predictors of voice disorders in thyroidectomy patients without RLN injury. Methods: Our [...] Read more.
Background: Thyroidectomy, a surgical procedure for thyroid disorders, is associated with postoperative voice changes, even in cases without recurrent laryngeal nerve (RLN) injury. Our study evaluates the prevalence and predictors of voice disorders in thyroidectomy patients without RLN injury. Methods: Our single-center prospective study at the University Hospital Center Zagreb included 243 patients, with pre- and postoperative voice evaluations using acoustic analysis and videostroboscopy. Logistic regression, chi-square, MANOVA, and non-parametric tests assessed the impact of surgical, sociodemographic, and lifestyle factors. Results: The study analyzed 243 participants (141 lobectomy, 102 total thyroidectomy). Postoperative voice disorders occurred in 200 patients (100 lobectomy, 100 total thyroidectomy); 43 (17.7%) experienced no voice disorders. Significant associations were observed for surgery type (χ2 = 29.88, p < 0.001), with total thyroidectomy having higher risk, surgery duration (χ2 = 16.40, p < 0.001), thyroid volume (χ2 = 4.24, p = 0.045), and BMI (χ2 = 8.97, p = 0.011). Gender and age showed no significant correlation. Acoustic parameters differed significantly, with lobectomy patients showing better intensity, jitter, and shimmer values across postoperative measurements. Logistic regression identified surgery type (Exp(B) = 16.533, p = 0.001) and thyroid volume (Exp(B) = 2.335, p = 0.023) as predictors of voice disorders, achieving 82.7% classification accuracy. Multivariate analysis confirmed gender and surgery duration as significant contributors. Surgery duration exceeding 90 min and enlarged thyroid volume negatively influenced outcomes. Significant acoustic differences were also linked to BMI categories, with obese participants exhibiting poorer parameters, particularly shimmer and jitter. Conclusions: Surgery type, thyroid volume, BMI, and surgery duration are most likely significant predictors of postoperative voice disorders. Full article
(This article belongs to the Special Issue Diagnosis and Management of Thyroid Disorders)
20 pages, 13806 KiB  
Article
Application of Mesenchymal Stem Cell-Derived Schwann Cell-like Cells Spared Neuromuscular Junctions and Enhanced Functional Recovery After Peripheral Nerve Injury
by Yu Hwa Nam, Ji-Sup Kim, Yoonji Yum, Juhee Yoon, Hyeryung Song, Ho-Jin Kim, Jaeseung Lim, Saeyoung Park and Sung-Chul Jung
Cells 2024, 13(24), 2137; https://doi.org/10.3390/cells13242137 - 23 Dec 2024
Viewed by 519
Abstract
In general, the nerve cells of the peripheral nervous system regenerate normally within a certain period after the physical damage of their axon. However, when peripheral nerves are transected by trauma or tissue extraction for cancer treatment, spontaneous nerve regeneration cannot occur. Therefore, [...] Read more.
In general, the nerve cells of the peripheral nervous system regenerate normally within a certain period after the physical damage of their axon. However, when peripheral nerves are transected by trauma or tissue extraction for cancer treatment, spontaneous nerve regeneration cannot occur. Therefore, it is necessary to perform microsurgery to connect the transected nerve directly or insert a nerve conduit to connect it. In this study, we applied human tonsillar mesenchymal stem cell (TMSC)-derived Schwann cell-like cells (TMSC-SCs) to facilitate nerve regeneration and prevent muscle atrophy after neurorrhaphy. The TMSC-SCs were manufactured in a good manufacturing practice facility and termed neuronal regeneration-promoting cells (NRPCs). A rat model of peripheral nerve injury (PNI) was generated and a mixture of NRPCs and fibrin glue was transplanted into the injured nerve after neurorrhaphy. The application of NRPCs and fibrin glue led to the efficient induction of sciatic nerve regeneration, with the sparing of gastrocnemius muscles and neuromuscular junctions. This sparing effect of NRPCs toward neuromuscular junctions might prevent muscle atrophy after neurorrhaphy. These results suggest that a mixture of NRPCs and fibrin glue may be a therapeutic candidate to enable peripheral nerve and muscle regeneration in the context of neurorrhaphy in patients with PNI. Full article
(This article belongs to the Special Issue The Role of Adipose-Derived Stem Cells in Neural Regeneration)
Show Figures

Figure 1

Figure 1
<p>Differentiation of NRPCs derived from TMSCs and comparison of the expression of peripheral nerve regeneration-related markers. TMSCs (<b>A</b>) and NRPCs (<b>B</b>) were observed under an inverted microscope. The scale bars indicate 200 μm. Comparison of the expression of peripheral nerve regeneration-related markers, including BDNF (<b>C</b>), GDNF (<b>D</b>), HGF (<b>E</b>), NRG1 (<b>F</b>), NGF (<b>G</b>), and NTF3 (<b>H</b>), in TMSCs and NRPCs, as assessed by WB. The expression levels were analyzed quantitatively using Image J software (Version 1.49) and normalized to that of GAPDH. Student’s <span class="html-italic">t</span>-test was performed to analyze and compare TMSCs and NRPCs. The data are presented as the mean ± SEM of three independent experiments (* <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). TMSCs, tonsil-derived mesenchymal stem cells; NRPCs, neuronal regeneration-promoting cells; WB, Western blotting; BDNF, brain-derived neurotrophic factor; GDNF, glial cell-derived neurotrophic factor; HGF, hepatocyte growth factor; NRG1, neuregulin 1; NGF, nerve growth factor; and NTF3, neurotrophin3.</p>
Full article ">Figure 2
<p>Observation of the myelination of the sciatic nerve of PNI rats using TEM. (<b>A</b>) Representative images of sciatic nerve cross-sections prepared at 2, 4, and 8 weeks after treatment (2 W, 4 W, and 8 W). The observation was not possible in the I group. Original magnification, 5000×. The axons in the only 8 W images were measured, followed by the quantification of the myelin thickness, indicating recovery of the sciatic nerve, based on the G-ratio (<b>B</b>) and the ratio of myelinated nerve fibers in the sciatic nerve (<b>C</b>). (<b>D</b>) Distribution of myelinated axons classified at 2 μm intervals between groups altered by NRPC application. Two-way ANOVA and Holm–Šídák’s multiple comparison test (row factor (number of axons per 2 μm diameter); column factor (group): (* <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; and **** <span class="html-italic">p</span> &lt; 0.0001)). PNI, peripheral nerve injury; TEM, transmission electron microscopy; NRPCs, neuronal regeneration-promoting cells; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + NRPC-L, neurorrhaphy + fibrin glue + NRPC-low; N + FG + NRPC-H, neurorrhaphy + fibrin glue + NRPC-high; W, wild type; and N/A, not applicable.</p>
Full article ">Figure 3
<p>Confirmation of neural function recovery in PNI rats after NRPC application by NSC analysis. (<b>A</b>) Original graph of the NCS results at 4, 8, and 12 weeks (4 W, 8 W, and 12 W) after treatment. The distal CMAP and NCV values measured from the NCS results were quantified and compared between the groups at 4 W (<b>B</b>,<b>C</b>), 8 W (<b>D</b>,<b>E</b>), and 12 W (<b>F</b>,<b>G</b>), respectively. One-way ANOVA and Tukey’s post hoc test were performed for comparison between groups. The data are presented as the mean ± SEM (* <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). PNI, peripheral nerve injury; NRPCs, neuronal regeneration-promoting cells; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + NRPC-L, neurorrhaphy + fibrin glue + NRPC-low; N + FG + NRPC-H, neurorrhaphy + fibrin glue + NRPC-high; W, wild type; NCS, nerve conduction study; CMAP, compound muscle action potential; and NCV, nerve conduction velocity.</p>
Full article ">Figure 4
<p>Axon regeneration and myelin formation in sciatic nerves in PNI rats treated with NRPCs. IHC staining for MBP (green) and NF-H (red) was performed at 6 (<b>A</b>) and 12 (<b>C</b>) weeks after NRPC application. The scale bars indicate 50 μm. Graph quantifying myelination of axons using IHC images at 6 (<b>B</b>) and 12 (<b>D</b>) weeks after NRPC application. One-way ANOVA and Tukey’s post hoc test were performed for comparison between groups. The data are presented as the mean ± SEM of three independent experiments (* <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). PNI, peripheral nerve injury; NRPCs, neuronal regeneration-promoting cells; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + NRPC-L, neurorrhaphy + fibrin glue + NRPC-low; N + FG + NRPC-H, neurorrhaphy + fibrin glue + NRPC-high; W, wild type; IHC, immunohistochemistry; MBP, myelin basic protein; and NF-H, neurofilament heavy.</p>
Full article ">Figure 5
<p>Confirmation of gastrocnemius muscle regeneration after NRPC application in PNI rats using H&amp;E staining. Histological changes in the gastrocnemius muscle were determined at 6 (<b>A</b>) and 12 (<b>B</b>) weeks after treatment. The image of H&amp;E staining was photographed under a microscope. White stars indicate big rounded fiber, and yellow circles indicate necrosis myofiber pattern. The scale bars indicate 50 μm. H&amp;E, hematoxylin and eosin; NRPCs, neuronal regeneration-promoting cells; PNI, peripheral nerve injury; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + NRPC-L, neurorrhaphy + fibrin glue + NRPC-low; N + FG + NRPC-H, neurorrhaphy + fibrin glue + NRPC-high; and W, wild type.</p>
Full article ">Figure 6
<p>Confirmation of gastrocnemius regeneration in PNI rats at 12 weeks after NRPC application. Immunohistochemistry tissue staining for skeletal muscle-related makers, such as MYH1E (green), MYH8 (green), and laminin (red). The cross-sectioned slides of the muscles were double-stained with MYH1E and laminin (<b>A</b>) or MYH8 and laminin (<b>D</b>), and those images were photographed and merged. All cells were counterstained with DAPI (blue). The scale bars indicate 100 μm. The expression levels of MYH1E and laminin (<b>B</b>,<b>C</b>) or MYH8 and laminin (<b>E</b>,<b>F</b>) were compared by quantification with DAPI using Image J software (Version 1.49). One-way ANOVA and Tukey’s post hoc test were performed for comparison between groups. The data are presented as the mean ± SEM of three independent experiments (* <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). PNI, peripheral nerve injury; NRPCs, neuronal regeneration-promoting cells; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + NRPC-L, neurorrhaphy + fibrin glue + NRPC-low; N + FG + NRPC-H, neurorrhaphy + fibrin glue + NRPC-high; W, wild type; MYH1E, myosin heavy chain 1 E; MYH8, myosin heavy chain 8; and DAPI, 4′,6-diamidino-2-phenylindole dihydrochloride.</p>
Full article ">Figure 7
<p>Expression of HGF and c-Met in PNI rat models by NRPC application. (<b>A</b>) Expression of HGF and c-Met in PNI rat nerves, as assessed by WB. The expression of HGF (<b>B</b>) and c-Met (<b>C</b>) was quantified using Image J software (Version 1.49), and its level was normalized to that of GAPDH. One-way ANOVA and Tukey’s post hoc test were performed for comparison between groups. The data are presented as the mean ± SEM (* <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). HGF, hepatocyte growth factor; c-Met, cellular mesenchymal–epithelial transition factor; PNI, peripheral nerve injury; NRPCs, neuronal regeneration-promoting cells; WB, Western blotting; TMSCs, tonsil-derived mesenchymal stem cells; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + NRPC-L, neurorrhaphy + fibrin glue + NRPC-low; N + FG + NRPC-H, neurorrhaphy + fibrin glue + NRPC-high; and W, wild type.</p>
Full article ">Figure 8
<p>Neuromuscular junction analysis in PNI rat models by NRPC application. (<b>A</b>) To confirm α-BTX (green) expression in muscle endplates, the samples were cryosectioned, stained, and observed using a slide scanner. The scale bar represents 100 μm. (<b>B</b>) The morphology of NMJs was observed by α-BTX (green) and NF-H (red) expression at high magnification using a confocal microscope. The scale bar represents 20 μm. NRPCs, neuronal regeneration-promoting cells; PNI, peripheral nerve injury; IHC, immunohistochemistry; TMSCs, tonsil-derived mesenchymal stem cells; I, injury; N, neurorrhaphy; N + FG, neurorrhaphy + fibrin glue; N + FG + TMSC, neurorrhaphy + fibrin glue + TMSC; N + FG + NRPC, neurorrhaphy + fibrin glue + NRPC; W, wild type; α-BTX, alpha-bungarotoxin; NMJ, neuromuscular junctions; and NF-H, neurofilament heavy.</p>
Full article ">Scheme 1
<p>Experimental scheme of cell transplantation and schedule of the animal experiment. (<b>A</b>) After producing the PNI rat model via sciatic nerve transection (<b>a</b>), neurorrhaphy was performed on the transected nerve (<b>b</b>), the background material was placed under the nerve (<b>c</b>), and a mixture of fibrin glue and NRPCs was transplanted (<b>d</b>), wrapped with the background material (<b>e</b>) and hardened for 1 min; then, the background material was removed (<b>f</b>). (<b>B</b>) Schematic diagram of the experiment used for the assessment of efficacy after the application of NRPCs to PNI rats. PNI, peripheral nerve injury; NRPCs, neuronal regeneration-promoting cells; TEM, transmission electron microscopy; NCS, nerve conduction study; and IHC, immunohistochemistry.</p>
Full article ">
10 pages, 1946 KiB  
Article
Prediction Model for Sciatic Nerve Procedures: A Cross-Sectional Study
by Isabel Minguez-Esteban, Ángel González-de-la-Flor, Jorge Hugo Villafañe, Juan Antonio Valera-Calero, Gustavo Plaza-Manzano, Pedro Belón-Pérez and Carlos Romero-Morales
J. Clin. Med. 2024, 13(24), 7851; https://doi.org/10.3390/jcm13247851 - 23 Dec 2024
Viewed by 398
Abstract
Objectives: We aimed to create a predictive model to estimate sciatic nerve depth using anthropometric and demographic data to enhance safety and precession in needle-based interventions. Setting: The study was conducted at Universidad Europea de Madrid, Spain. Methods: A Cross-sectional observational study was [...] Read more.
Objectives: We aimed to create a predictive model to estimate sciatic nerve depth using anthropometric and demographic data to enhance safety and precession in needle-based interventions. Setting: The study was conducted at Universidad Europea de Madrid, Spain. Methods: A Cross-sectional observational study was carried out between January and April 2024. The study included fifty volunteers aged 18–45 years, without any muscle tone affections, lower limb asymmetries, or history of lower limb surgeries. Demographic and anthropometric data were collected, including sex, age, height, weight, BMI, and leg length measure and thigh circumference at specific points. The sciatic nerve depth was measured using ultrasound imaging under the gluteal fold and in the posterior middle third of the thigh. Results: Correlation analysis revealed significant associations between thigh circumference at the proximal and middle third and sciatic nerve depth. A multiple linear regression model identified that the proximal thigh circumference was a significant predictor of sciatic nerve depth, explaining 44.5% of the variance. The variance increased to 49.7% when gender was added. The depth of the sciatic nerve in the middle third explained 38.2% of the variance. And the inclusion of gender in the model explained 40.8% of the variance for the middle third. Conclusions: This study identify significant predictors such as the thigh girth at the proximal and mid-third levels, gender, and the BMI. These findings suggest that clinicians can use these anthropometric measurements to estimate sciatic nerve depth more accurately, reducing the risk of accidental nerve injury and improve the precision and safety of needling procedures during invasive procedures. Full article
Show Figures

Figure 1

Figure 1
<p>SN: sciatic nerve; CT: conjoint tendon; BF: biceps femoris; ST: semitendinous; SMT: semimembranosus tendon; AM: adductor magnus.</p>
Full article ">Figure 2
<p>SN: sciatic nerve; CT: conjoint tendon; BF: biceps femoris; ST: semitendinous; SMT: semimembranosus tendon; AM: adductor magnus.</p>
Full article ">Figure 3
<p>Scatter plot showcasing the relationship between observed values (Y-axis) and predicted values (X-axis) for the distance from the skin surface to the sciatic nerve (crossmarks, measured in cm) at the proximal third (<b>A</b>) and mid-third (<b>B</b>). The solid black line depicts the linear regression model, while the dashed lines represent the 95% prediction interval boundaries.</p>
Full article ">
13 pages, 35081 KiB  
Article
Comparison of the Effects of Perineural and Intraperitoneal Ozone Therapy on Nerve Healing in an Experimental Sciatic Nerve Injury Model
by Burcu Ayık, Abdullah Ortadeveci, Fulya Bakılan, Dilek Burukoğlu Dönmez, Semih Öz, Cengiz Bal, Hilmi Özden and Onur Armağan
Medicina 2024, 60(12), 2097; https://doi.org/10.3390/medicina60122097 - 21 Dec 2024
Viewed by 467
Abstract
Background and Objectives: The aim was to evaluate nerve healing using immunohistochemical, histological, and functional techniques and to compare the effects of two different therapeutic ozone application methods by perineural and intraperitoneal ozone treatment in rats with a crush injury model of sciatic [...] Read more.
Background and Objectives: The aim was to evaluate nerve healing using immunohistochemical, histological, and functional techniques and to compare the effects of two different therapeutic ozone application methods by perineural and intraperitoneal ozone treatment in rats with a crush injury model of sciatic nerve. Materials and Methods: Forty male Sprague Dawley rats were divided into four subgroups of ten rats each: (1) Control group: The left sciatic nerve incised and closed without crush injury, no treatment; (2) Paralyzed group: Crush injury to the left sciatic nerve, no treatment; (3) Perineural ozone group: Crush injury to the left sciatic nerve, treated with perineural ozone therapy; (4) Intraperitoneal ozone group: Crush injury to the left sciatic nerve, treated with intraperitoneal ozone therapy. The treatments were administered for a 14-day period. Hematoxylin and eosin (H&E) and toluidine blue staining were used for histological examination; TUNEL staining was used for immunohistochemical examination. Pinch test and rotarod performance assessment were utilized for functional evaluation. Results: The pinch test scores showed significant improvement in perineural and intraperitoneal ozone treatment groups after treatment (p < 0.001 and p = 0.003, respectively). The scores of myelin degeneration, vascular congestion, vascular wall thickness, inflammation, and toluidine blue and TUNEL staining were significantly lower in both ozone treatment groups compared to the paralyzed group (p < 0.001). Vascular wall thickness scores were significantly higher in the perineural ozone group compared to the control and intraperitoneal ozone groups (p = 0.004 and p = 0.013, respectively). The Schwann cell proliferation scores were significantly higher in the perineural ozone group compared to the control group and intraperitoneal ozone groups (p < 0.001). Conclusions: Both applications of ozone therapy accelerated the healing of nerve regeneration, reduced inflammation and apoptosis based on histopathological results, and enhanced nerve function in rats with sciatic nerve injury. Perineural ozone therapy has been demonstrated to be an efficacious alternative to systemic ozone treatments in the management of sciatic nerve injury. Further studies are needed to determine optimal ozone dosage and administration protocols for the treatment of nerve injury. Full article
(This article belongs to the Special Issue New Insights into Neurodevelopmental Biology and Disorders)
Show Figures

Figure 1

Figure 1
<p>Study flow chart.</p>
Full article ">Figure 2
<p>Region of the sciatic nerve dissection.</p>
Full article ">Figure 3
<p>Light microscopy examination images of sciatic nerve specimens from each group. ((<b>A1</b>–<b>A3</b>): Control group, (<b>B1–B3</b>): Paralyzed group, (<b>C1</b>–<b>C3</b>): Perineural ozone group, (<b>D1</b>–<b>D3</b>): Intraperitoneal ozone group.) (ep: epineurium, pe: perineurium, en: endoneurium, inf: inflammation, v: vessel, Hematoxylin and eosin (H&amp;E), scale bar: 200 µm-×10, 100 µm-×20, 50 µm-×40.) Myelinated axon structures (<span class="html-fig-inline" id="medicina-60-02097-i001"><img alt="Medicina 60 02097 i001" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i001.png"/></span>), myelinated axon structures observed in degenerated vacuolar structure (<span class="html-fig-inline" id="medicina-60-02097-i002"><img alt="Medicina 60 02097 i002" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i002.png"/></span>), vascular congestion (<span class="html-fig-inline" id="medicina-60-02097-i003"><img alt="Medicina 60 02097 i003" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i003.png"/></span>), vascular wall thickening (<span class="html-fig-inline" id="medicina-60-02097-i004"><img alt="Medicina 60 02097 i004" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i004.png"/></span>), and proliferating Schwann cells (<span class="html-fig-inline" id="medicina-60-02097-i005"><img alt="Medicina 60 02097 i005" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i005.png"/></span>). Light microscopy examination of the sciatic nerve specimens taken from the control group after hematoxylin–eosin staining showed a normal histological structure characterized by intact myelinated axons and vascular structures. In the paralyzed group, the light microscopy examination revealed degenerated vacuolar structures, vascular congestion, vascular wall thickening, and inflammation in myelinated axon structures. In the perineural ozone group, near normal myelinated axon structures as well as decreased vascular wall thickening and proliferated Schwann cells were observed in some areas. In the intraperitoneal ozone group, although a small number of vascular congestion was observed in some areas, it revealed a histologic structure close to normal in general evaluation.</p>
Full article ">Figure 4
<p>Light microscopy images of toluidine blue-stained preparations of sciatic nerve specimens from each group. ((<b>A1</b>–<b>A3</b>): Control group, (<b>B1</b>–<b>B3</b>): Paralyzed group, (<b>C1</b>–<b>C3</b>): Perineural ozone group, (<b>D1</b>–<b>D3</b>): Intraperitoneal ozone group) (Toluidine blue, scale bar: 200 µm-×10, 100 µm-×20, 50 µm-×40.) Mast cells (<span class="html-fig-inline" id="medicina-60-02097-i006"><img alt="Medicina 60 02097 i006" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i006.png"/></span>), myelinated axon structures observed in degenerated vacuolar structure (<span class="html-fig-inline" id="medicina-60-02097-i007"><img alt="Medicina 60 02097 i007" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i007.png"/></span>), normal myelinated axon structures (<span class="html-fig-inline" id="medicina-60-02097-i008"><img alt="Medicina 60 02097 i008" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i008.png"/></span>), and proliferating Schwann cells (<span class="html-fig-inline" id="medicina-60-02097-i009"><img alt="Medicina 60 02097 i009" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i009.png"/></span>). The light microscopy examination of toluidine blue-stained sciatic nerve specimens revealed a normal histological structure characterized by mast cells, myelinated axon structures, and vascular formations in the control group. In the paralyzed group, mast cells, degenerated vacuolar structures in myelinated axon structures, and few proliferating Schwann cells were detected. In the perineural and intraperitoneal ozone groups, mast cells and myelinated axon structures, reduced degenerated myelinated axon structures, and proliferating Schwann cells were detected in some areas.</p>
Full article ">Figure 5
<p>Immunohistochemical examinations following TUNEL staining of sciatic nerve specimens from each group. ((<b>A1</b>–<b>A3</b>): Control group, (<b>B1</b>–<b>B3</b>): Paralyzed group, (<b>C1</b>–<b>C3</b>): Perineural ozone group, (<b>D1</b>–<b>D3</b>): Intraperitoneal ozone group.) (TUNEL, scale bar: 200 µm-×10, 100 µm-×20, 50 µm-×40.) TUNEL staining (<span class="html-fig-inline" id="medicina-60-02097-i010"><img alt="Medicina 60 02097 i010" src="/medicina/medicina-60-02097/article_deploy/html/images/medicina-60-02097-i010.png"/></span>). The light microscopy examination of the specimens subjected to TUNEL staining revealed minimal positive staining in the control group, advanced positive staining in the paralyzed group, and moderate positive staining in both ozone groups.</p>
Full article ">
27 pages, 4435 KiB  
Article
Remote Ischemic Post-Conditioning (RIC) Mediates Anti-Inflammatory Signaling via Myeloid AMPKα1 in Murine Traumatic Optic Neuropathy (TON)
by Naseem Akhter, Jessica Contreras, Mairaj A. Ansari, Andrew F. Ducruet, Md Nasrul Hoda, Abdullah S. Ahmad, Laxman D. Gangwani, Kanchan Bhatia and Saif Ahmad
Int. J. Mol. Sci. 2024, 25(24), 13626; https://doi.org/10.3390/ijms252413626 - 19 Dec 2024
Viewed by 472
Abstract
Traumatic optic neuropathy (TON) has been regarded a vision-threatening condition caused by either ocular or blunt/penetrating head trauma, which is characterized by direct or indirect TON. Injury happens during sports, vehicle accidents and mainly in military war and combat exposure. Earlier, we have [...] Read more.
Traumatic optic neuropathy (TON) has been regarded a vision-threatening condition caused by either ocular or blunt/penetrating head trauma, which is characterized by direct or indirect TON. Injury happens during sports, vehicle accidents and mainly in military war and combat exposure. Earlier, we have demonstrated that remote ischemic post-conditioning (RIC) therapy is protective in TON, and here we report that AMPKα1 activation is crucial. AMPKα1 is the catalytic subunit of the heterotrimeric enzyme AMPK, the master regulator of cellular energetics and metabolism. The α1 isoform predominates in immune cells including macrophages (Mφs). Myeloid-specific AMPKα1 KO mice were generated by crossing AMPKα1Flox/Flox and LysMcre to carry out the study. We induced TON in mice by using a controlled impact system. Mice (mixed sex) were randomized in six experimental groups for Sham (mock); Sham (RIC); AMPKα1F/F (TON); AMPKα1F/F (TON+RIC); AMPKα1F/F LysMCre (TON); AMPKα1F/F LysMCre (TON+RIC). RIC therapy was given every day (5–7 days following TON). Data were generated by using Western blotting (pAMPKα1, ICAM1, Brn3 and GAP43), immunofluorescence (pAMPKα1, cd11b, TMEM119 and ICAM1), flow cytometry (CD11b, F4/80, CD68, CD206, IL-10 and LY6G), ELISA (TNF-α and IL-10) and transmission electron microscopy (TEM, for demyelination and axonal degeneration), and retinal oxygenation was measured by a Unisense sensor system. First, we observed retinal morphology with funduscopic images and found TON has vascular inflammation. H&E staining data suggested that TON increased retinal inflammation and RIC attenuates retinal ganglion cell death. Immunofluorescence and Western blot data showed increased microglial activation and decreased retinal ganglion cell (RGCs) marker Brn3 and axonal regeneration marker GAP43 expression in the TON [AMPKα1F/F] vs. Sham group, but TON+RIC [AMPKα1F/F] attenuated the expression level of these markers. Interestingly, higher microglia activation was observed in the myeloid AMPKα1F/F KO group following TON, and RIC therapy did not attenuate microglial expression. Flow cytometry, ELISA and retinal tissue oxygen data revealed that RIC therapy significantly reduced the pro-inflammatory signaling markers, increased anti-inflammatory macrophage polarization and improved oxygen level in the TON+RIC [AMPKα1F/F] group; however, RIC therapy did not reduce inflammatory signaling activation in the myeloid AMPKα1 KO mice. The transmission electron microscopy (TEM) data of the optic nerve showed increased demyelination and axonal degeneration in the TON [AMPKα1F/F] group, and RIC improved the myelination process in TON [AMPKα1F/F], but RIC had no significant effect in the AMPKα1 KO mice. The myeloid AMPKα1c deletion attenuated RIC induced anti-inflammatory macrophage polarization, and that suggests a molecular link between RIC and immune activation. Overall, these data suggest that RIC therapy provided protection against inflammation and neurodegeneration via myeloid AMPKα1 activation, but the deletion of myeloid AMPKα1 is not protective in TON. Further investigation of RIC and AMPKα1 signaling is warranted in TON. Full article
(This article belongs to the Special Issue New Therapeutic Targets for Neuroinflammation and Neurodegeneration)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Representative in vivo funduscopic fluorescein image from C56BL/6 mice showing inflammation in blood vessels in TON as compared with control eye. Intravenous fluorescein angiography of the mouse retina shows poor perfusion through attenuated vasculature (due to progression of the retinal degeneration) following TON. (<b>B</b>) H&amp;E data showed increased neuronal cell death in ganglion cell layer in TON compared with control. However, the neuronal cell death is prevented with RIC treatment. Fluorescein angiography imaging (<b>A</b>) was captured within 5 mins of fluorescein dye injection through tail vein.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Immunofluorescence staining showed microglial marker TMEM119 expression in mouse retina. TON (with AMPK) increases microglial activation, and RIC downregulated significantly. Myeloid pAMPKα1 KO group showed heightened microglial activation; notably, RIC demonstrated no significant effects. Florescence color intensity was measured by Image J software (NIH, <a href="https://imagej.net/ij/" target="_blank">https://imagej.net/ij/</a>). White boxes show the TMEM119 expression in inner nuclear layer (INL) and GCL (ganglion cell layer) region of mouse eye. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>. (<b>C</b>–<b>I</b>) Representative pseudocolor and histograms of flow cytometry show the gating strategy for microglia/macrophages (CD11b+_F4/80+) and CD68+ and CD206+ expressing microglia in blood. Bar graph summarizing the cell counts of microglia (M1/M2) in the blood after 5 days of TON. Red, TMEM119 (activated microglial marker); Blue, DAPI. We used 6 experimental groups, Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysMCre (TON); AMPKα1<sup>F/F</sup> LysMCre (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>,<b>B</b>) Immunofluorescence staining showed microglial marker TMEM119 expression in mouse retina. TON (with AMPK) increases microglial activation, and RIC downregulated significantly. Myeloid pAMPKα1 KO group showed heightened microglial activation; notably, RIC demonstrated no significant effects. Florescence color intensity was measured by Image J software (NIH, <a href="https://imagej.net/ij/" target="_blank">https://imagej.net/ij/</a>). White boxes show the TMEM119 expression in inner nuclear layer (INL) and GCL (ganglion cell layer) region of mouse eye. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>. (<b>C</b>–<b>I</b>) Representative pseudocolor and histograms of flow cytometry show the gating strategy for microglia/macrophages (CD11b+_F4/80+) and CD68+ and CD206+ expressing microglia in blood. Bar graph summarizing the cell counts of microglia (M1/M2) in the blood after 5 days of TON. Red, TMEM119 (activated microglial marker); Blue, DAPI. We used 6 experimental groups, Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysMCre (TON); AMPKα1<sup>F/F</sup> LysMCre (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 3
<p>Effect of RIC on IL10 and neutrophil expression following TON. (<b>A</b>,<b>B</b>,<b>D</b>,<b>F</b>) Representative pseudocolor and histograms of flow cytometry show the gating strategy for microglia/macrophages (CD11b+_IL10+, F4/80+_IL10+ and CD68+_IL10+) and CD68+_LY6G+-expressing neutrophils in blood. (<b>C</b>,<b>E</b>,<b>G</b>,<b>H</b>) Representative bar graph summarizing the cell counts of IL10+ and Ly6G+ in the blood after 5 days of TON. Six experimental groups included Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysMCre (TON); AMPKα1<sup>F/F</sup> LysMCre (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). ** <span class="html-italic">p</span> &lt; 0.01.*** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 3 Cont.
<p>Effect of RIC on IL10 and neutrophil expression following TON. (<b>A</b>,<b>B</b>,<b>D</b>,<b>F</b>) Representative pseudocolor and histograms of flow cytometry show the gating strategy for microglia/macrophages (CD11b+_IL10+, F4/80+_IL10+ and CD68+_IL10+) and CD68+_LY6G+-expressing neutrophils in blood. (<b>C</b>,<b>E</b>,<b>G</b>,<b>H</b>) Representative bar graph summarizing the cell counts of IL10+ and Ly6G+ in the blood after 5 days of TON. Six experimental groups included Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysMCre (TON); AMPKα1<sup>F/F</sup> LysMCre (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). ** <span class="html-italic">p</span> &lt; 0.01.*** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 4
<p>The effect of RIC on TON induced pro-inflammatory signaling. (<b>A</b>,<b>B</b>) ELISA results in blood plasma showing TNF and IL10 expression. Fluorescence color intensity was measured by Image J software. We used 6 experimental group, Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysM<sup>Cre</sup> (TON); AMPKα1<sup>F/F</sup> LysM<sup>Cre</sup> (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <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, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 5
<p>Effect of RIC on TON induced pro-inflammatory signaling. (<b>A</b>,<b>B</b>) The effect of RIC on ICAM-1 expression assessed by immunofluorescence and (<b>C</b>,<b>D</b>) ICAM1 Protein expression was checked by Western blot. Fluorescence color intensity as well as western blot band intensity was measured by Image J software (NIH, <a href="https://imagej.net/ij/" target="_blank">https://imagej.net/ij/</a>). We used 6 experimental groups, Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysM<sup>Cre</sup> (TON); AMPKα1<sup>F/F</sup> LysM<sup>Cre</sup> (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <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, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. Scale bar 50 μm. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 5 Cont.
<p>Effect of RIC on TON induced pro-inflammatory signaling. (<b>A</b>,<b>B</b>) The effect of RIC on ICAM-1 expression assessed by immunofluorescence and (<b>C</b>,<b>D</b>) ICAM1 Protein expression was checked by Western blot. Fluorescence color intensity as well as western blot band intensity was measured by Image J software (NIH, <a href="https://imagej.net/ij/" target="_blank">https://imagej.net/ij/</a>). We used 6 experimental groups, Sham (mock); Sham (RIC); AMPKα1<sup>F/F</sup> (TON); AMPKα1<sup>F/F</sup> (TON+RIC); AMPKα1<sup>F/F</sup> LysM<sup>Cre</sup> (TON); AMPKα1<sup>F/F</sup> LysM<sup>Cre</sup> (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <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, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. Scale bar 50 μm. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 6
<p>(<b>A</b>–<b>D</b>) Effect of RIC therapy on retinal oxygenation in TON. Oxygen levels were analyzed with UniSense sensor system (Sweden). We used 6 experimental groups, Sham (mock); Sham (RIC); AMPKα1F/F (TON); AMPKα1F/F (TON+RIC); AMPKα1F/F LysM<sup>Cre</sup> (TON); AMPKα1F/F LysM<sup>Cre</sup> (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <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, **** <span class="html-italic">p</span> &lt; 0.0001. ns, non-significant. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 7
<p>(<b>A</b>–<b>D</b>) Effect of RIC therapy on TON retina. Western blot analysis demonstrated significant changes in protein expression level of Brn3a and GAP43 between TON+RIC and TON group. Densitometry analysis was carried out by Image J software (NIH, <a href="https://imagej.net/ij/" target="_blank">https://imagej.net/ij/</a>). We used 4 experimental groups, AMPKα1F/F (TON); AMPKα1F/F (TON+RIC); AMPKα1F/F LysM<sup>Cre</sup> (TON); AMPKα1F/F LysM<sup>Cre</sup> (TON+RIC). Differences among experimental groups were determined by analysis of variance (one-way ANOVA) followed by Newman–Keuls multiple comparison tests. The results represent the means ± SEM of fold changes (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns, non-significant.</p>
Full article ">Figure 8
<p>Representative ultrastructural features of axonal injury in traumatic optic neuropathy. Electron micrographs are taken across the longitudinal plane through the injury front and show a range of axoplasmic, axolemmal and myelin sheath abnormalities. RIC therapy attenuated this degenerating process in TON. We used 6 experimental groups, Sham (mock); Sham (RIC); AMPKα1F/F (TON); AMPKα1F/F (TON+RIC); AMPKα1F/F LysM<sup>Cre</sup> (TON); AMPKα1F/F LysM<sup>Cre</sup> (TON+RIC). Scale bar 4 μm. For Sham (mock) and Sham (RIC), both groups are regarded as AMPKα1<sup>F/F</sup>.</p>
Full article ">Figure 9
<p>Schematic representation demonstrating increased M1-type macrophages causing inflammation and demyelination of optic nerve (ON) in TON. Our hypothesis demonstrates that RIC therapy activates AMPKα1 to modulate macrophage polarization toward M2-type anti-inflammatory macrophages that protect demyelination of downregulated ON.</p>
Full article ">
21 pages, 3403 KiB  
Review
Coordinated Actions of Neurogenesis and Gliogenesis in Nerve Injury Repair and Neuroregeneration
by Mei-Yu Chen, Cheng-Yu Chi, Chiau-Wei Zheng, Chen-Hung Wang and Ing-Ming Chiu
Int. J. Transl. Med. 2024, 4(4), 810-830; https://doi.org/10.3390/ijtm4040053 - 19 Dec 2024
Viewed by 438
Abstract
The failure of endogenous repair mechanisms is a key characteristic of neurological diseases, leading to the inability to restore damaged nerves and resulting in functional impairments. Since the endogenously regenerative capacity of damaged nerves is limited, the enhancement of regenerative potential of quiescent [...] Read more.
The failure of endogenous repair mechanisms is a key characteristic of neurological diseases, leading to the inability to restore damaged nerves and resulting in functional impairments. Since the endogenously regenerative capacity of damaged nerves is limited, the enhancement of regenerative potential of quiescent neural stem cells (NSCs) presents as a therapeutic option for neural diseases. Our previous studies have shown exciting progress in treating sciatic nerve injury in mice and rats using NSCs in conjunction with neurotrophic factors such as fibroblast growth factor 1 (FGF1). Additionally, a recently discovered neurotrophic factor, IL12p80, has shown significant therapeutic effects in sciatic nerve injury repair via myelinating oligodendrocytes. IL12p80 induces oligodendrocyte differentiation from NSCs through phosphorylation of Stat3. Therefore, it might be possible to alleviate the myelination defects of oligodendrocytes in neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and even schizophrenia through the administration of IL12p80. These applications could shed light on IL12p80 and FGF1, not only in damaged nerve repair, but also in rectifying the oligodendrocytes’ defects in neurodegenerative diseases, such as ALS and MS. Finally, the synergistic effects of neurogenesis-induced FGF1 and myelination-induced IL12 might be able to supplant the need of NSCs for nerve repair and neuroregeneration. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Transcriptional regulation of endogenous FGF1 expression in different tissues. The human FGF1 gene structure is schematically presented with a scale (kbp). Exons –1A, –1B, –1C, and –1D are the alternative exons generated using promoters A, B, C, and D, respectively [<a href="#B96-ijtm-04-00053" class="html-bibr">96</a>].</p>
Full article ">Figure 2
<p>GFP fluorescence permits the isolation and purification of F1B-positive brain cells from F1B-Tag transgenic mice. F1B-Tag/F1B-GFP(+) and F1B-Tag/F1B-GFP(−) cells were separated via fluorescence-activated cell sorting. The F1B-GFP(+) cells possess remarkable neurosphere-forming activity when compared with F1B-GFP(−) [<a href="#B96-ijtm-04-00053" class="html-bibr">96</a>]. Furthermore, F1B-GFP(+) cells could differentiate into neurons, astroglial cells, and oligodendrocytes, demonstrating their multipotent capacities [<a href="#B102-ijtm-04-00053" class="html-bibr">102</a>]. Thus, F1B-positive brain cells from F1B-Tag transgenic mice showed self-renewal and multipotent capacities [<a href="#B96-ijtm-04-00053" class="html-bibr">96</a>,<a href="#B102-ijtm-04-00053" class="html-bibr">102</a>].</p>
Full article ">Figure 3
<p>Assessment of functional recovery via walking track analysis, using the rat’s footprint areas as indices. A brief description is as follows: Preoperatively, the rats were trained to walk down a 150 × 8 cm track in a darkened enclosure. The sciatic functional index (SFI), which assessed the functional muscle reinnervation, was calculated based on the walking track analysis using the following equation: SFI = −38.3(PLF) + 109.5(TSF) + 13.3(ITF) − 8.8, where PLF (print length function) = (experimental PL − normal PL)/normal PL, TSF (toe spread function) = (experimental TS − normal TS)/normal TS (1st to 5th Toe), and ITF (inter-median toe spread function) = (experimental IT − normal IT)/normal IT (2nd to 4th Toe) [<a href="#B110-ijtm-04-00053" class="html-bibr">110</a>]. The footprinted area in the walking track analysis was further scanned and recorded with an image analysis system (Image-Pro Lite, Media Cybernetics, Rockville, MD, USA). The ratio of the experimental foot area/normal foot area was analyzed. The degrees of repair could be quantitated using SFI analyses, as described in our publication [<a href="#B97-ijtm-04-00053" class="html-bibr">97</a>].</p>
Full article ">Figure 4
<p>Sciatic functional index analyses of rats with transected sciatic nerves and treated with GFP-positive NSCs using PLA-grooved nerve conduits with FGF1 and NSCs. Cn: rats repaired using conduits alone (<span style="color:blue">△</span>); Cn+NSCs: rats repaired using conduits with NSCs (<span style="color:red">○</span>); Cn+FGF1: rats repaired using conduits with FGF1 (<span style="color:blue">▲</span>); Cn+FGF1+NSCs: rats repaired using conduits with FGF1 and NSCs (<span style="color:red">●</span>). Four rats were used in each group. The Cn+FGF1+NSCs group shows better functional recovery than any of the other three groups. The results indicate that using the treatment comprising stem cells, FGF1, and conduits is the best strategy for sciatic nerve injury repair in rats [<a href="#B97-ijtm-04-00053" class="html-bibr">97</a>].</p>
Full article ">Figure 5
<p>Diameters of regenerated sciatic nerve were increased with the administering of IL12. Four mice were used in each group. Mouse IL12p80 increases the diameter of a regenerated nerve up to 4.5-fold when NSCs or NSCs+IL12p80 were incorporated in the conduits, from 65 µm to 189 µm and 295 µm, respectively, at the medial section of the regenerated nerve. Mouse sciatic nerve injury repaired using conduits alone (<span style="color:red">■</span>); using conduits with NSCs (<span style="color:green">■</span>); using conduits with NSCs and IL12p80 (<span style="color:blue">■</span>) [<a href="#B114-ijtm-04-00053" class="html-bibr">114</a>].</p>
Full article ">Figure 6
<p>Enhancement of nerve regeneration in the sciatic nerve injury mouse model through the implantation of PLA conduits with NSCs and human IL12p80. (<b>A</b>–<b>D</b>) Staining of tissue sections with hematoxylin and eosin (H&amp;E) was carried out for the measurements of the sizes of the regenerated sciatic nerve in mice. “P” marks the proximal site of the residual sciatic nerves in mice, while “D” marks the distal site (“P” and “D” are 3.0 mm apart). (<b>E</b>–<b>H</b>). Immunohistochemical staining using anti-NF200 antibody (green) and anti-PZ0 antibody (red). Nuclei were stained with DAPI (blue). NF200 and PZ0 are the markers for nerve fibers and myelinating Schwann cells, respectively. Four mice were used in each group. Scale bars: (<b>A</b>–<b>D</b>), 1.0 mm; (<b>E</b>–<b>H</b>), 200 µm [<a href="#B133-ijtm-04-00053" class="html-bibr">133</a>].</p>
Full article ">
9 pages, 1775 KiB  
Article
Evaluating Meniscus, Ligament and Soft Tissue Injury Using MRI in Tibial Plateau Fractures: A Tscherne Classification Approach
by Yong-Bum Joo, Young-Mo Kim, Young-Cheol Park, Soo-Hyeok Chae and Dong-Hwan Kim
Medicina 2024, 60(12), 2073; https://doi.org/10.3390/medicina60122073 - 17 Dec 2024
Viewed by 431
Abstract
Background and Objectives: This study investigated associated meniscus and ligament injuries in tibial plateau fractures using magnetic resonance imaging (MRI) and assessed soft tissue injuries in relation to the Schatzker classification and Tscherne classification. Materials and Methods: The data of 185 [...] Read more.
Background and Objectives: This study investigated associated meniscus and ligament injuries in tibial plateau fractures using magnetic resonance imaging (MRI) and assessed soft tissue injuries in relation to the Schatzker classification and Tscherne classification. Materials and Methods: The data of 185 patients who sustained tibial plateau fractures from January 2010 to April 2021 were retrospectively reviewed. Fractures were classified according to the Schatzker classification system. Soft-tissue injuries were assessed using the Tscherne classification. Menisci and ligaments were evaluated using preoperative MRI. Nerve injuries, compartment syndrome and wound problems were also assessed. The incidence of soft tissue injuries, as well as the relationship between the Schatzker and Tscherne classification systems, were analyzed. Results: Evidence of derangement of meniscus and ligament around the knee was found in 183 (98.9%) patients. The most common injury was a medial collateral ligament injury. The incidence of lateral collateral ligament injury, nerve injury, compartment syndrome and wound problem was higher in high-energy tibial plateau fractures. A tendency was observed between the Schatzker and the Tscherne classifications (p value < 0.001). Higher Tscherne grade was also associated with the incidence of posterior cruciate ligament injury, nerve injury and compartment syndrome. Conclusions: In tibial plateau fractures, soft tissue injuries were highly prevalent. High-energy fractures tended to exhibit higher Tscherne classification grades and showed an increased incidence of meniscus and ligament injuries. The Tscherne classification appears to be a helpful system for predicting soft tissue injuries in tibial plateau fractures. And preoperative MRI can be a helpful tool. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

Figure 1
<p>Flow diagram illustrating patient enrolment. MRI: Magnetic resonance imaging.</p>
Full article ">Figure 2
<p>The Tscherne classification categorizes the extent of soft tissue injury associated with fracture trauma. (<b>A</b>) grade 0; none to minimal soft tissue damage, (<b>B</b>) grade 1; superficial abrasion or skin contusion, (<b>C</b>) grade 2; deep abrasion or muscle contusion, (<b>D</b>) grade 3; extensive skin contusion, crush injury with severe damage to underlying muscle, Morel-Lavallee lesion and/or vascular injury.</p>
Full article ">Figure 3
<p>A 35-year-old male. (<b>A</b>,<b>B</b>) Preoperative anteroposterior and lateral plain X-ray shows a left proximal tibial fracture. (<b>C</b>–<b>F</b>) 3D CT shows bicondylar fracture including metaphyseal-diaphyseal discontinuity. This type is Schatzker type VI and high-energy fracture.</p>
Full article ">Figure 4
<p>Same patient with <a href="#medicina-60-02073-f002" class="html-fig">Figure 2</a> (<b>A</b>) Preoperative MRI shows soft tissue injury of the Tscherne classification grade 3. (<b>B</b>) On day 1 after injury, the patient’s lower leg was severely swollen and showed hemorrhagic fracture blisters.</p>
Full article ">Figure 5
<p>Same patient with <a href="#medicina-60-02073-f002" class="html-fig">Figure 2</a> (<b>A</b>,<b>B</b>) Temporary external fixation was done. (<b>C</b>,<b>D</b>) 20 days after external fixator application, dual locking compression plate fixation was done. (<b>E</b>,<b>F</b>) 18 months after plate application, bony union was seen and all metal materials except one broken screw were removed.</p>
Full article ">
32 pages, 18493 KiB  
Article
Discovery of Glucose Metabolism-Associated Genes in Neuropathic Pain: Insights from Bioinformatics
by Ying Yu, Yan-Ting Cheung and Chi-Wai Cheung
Int. J. Mol. Sci. 2024, 25(24), 13503; https://doi.org/10.3390/ijms252413503 - 17 Dec 2024
Viewed by 284
Abstract
Metabolic dysfunction has been demonstrated to contribute to diabetic pain, pointing towards a potential correlation between glucose metabolism and pain. To investigate the relationship between altered glucose metabolism and neuropathic pain, we compared samples from healthy subjects with those from intervertebral disc degeneration [...] Read more.
Metabolic dysfunction has been demonstrated to contribute to diabetic pain, pointing towards a potential correlation between glucose metabolism and pain. To investigate the relationship between altered glucose metabolism and neuropathic pain, we compared samples from healthy subjects with those from intervertebral disc degeneration (IVDD) patients, utilizing data from two public datasets. This led to the identification of 412 differentially expressed genes (DEG), of which 234 were upregulated and 178 were downregulated. Among these, three key genes (Ins, Igfbp3, Plod2) were found. Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated the enrichment of hub genes in pathways such as the positive regulation of the ErbB signaling pathway, monocyte activation, and response to reactive oxygen species; thereby suggesting a potential correlation between these biological pathways and pain sensation. Further analysis identified three key genes (Ins, Igfbp3, and Plod2), which showed significant correlations with immune cell infiltration, suggesting their roles in modulating pain through immune response. To validate our findings, quantitative real-time polymerase chain reaction (qPCR) analysis confirmed the expression levels of these genes in a partial sciatic nerve ligation (PSNL) model, and immunofluorescence studies demonstrated increased immune cell infiltration at the injury site. Behavioral assessments further corroborated pain hypersensitivity in neuropathic pain (NP) models. Our study sheds light on the molecular mechanisms underlying NP and aids the identification of potential therapeutic targets for future drug development. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Figure 1

Figure 1
<p>Identification of differentially expressed genes (DEGs) in neuropathic pain (NP) patients and healthy controls. (<b>A</b>) Principal component analysis (PCA) plot of the two datasets before batch effect correction, showing clear batch-related variability. (<b>B</b>) PCA plot of the two datasets after batch effect correction, demonstrating successful integration with reduced batch effects. (<b>C</b>) Volcano plot displaying all DEGs, with upregulated genes in red, downregulated genes in blue, and non-significant genes in gray. (<b>D</b>) Heatmap of the top 20 upregulated and downregulated DEGs, highlighting their differential expression between NP patients and healthy controls.</p>
Full article ">Figure 2
<p>Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of differentially expressed genes (DEGs). (<b>A</b>,<b>C</b>,<b>E</b>) Circular plots showing the top enriched GO terms for DEGs in the categories of biological process (BP) (<b>A</b>), cellular component (CC) (<b>C</b>), and molecular function (MF) (<b>E</b>). Each plot connects DEGs to their corresponding GO terms, with line colors reflecting the log fold change of the genes and circle sizes representing the number of associated genes for each term. (<b>B</b>,<b>D</b>,<b>F</b>) Dot plots summarizing the GO enrichment results for BP (<b>B</b>), CC (<b>D</b>), and MF (<b>F</b>). The x-axis indicates the enrichment score (−log10(<span class="html-italic">p</span>-value)), while dot colors represent <span class="html-italic">p</span>-value significance, and dot sizes correspond to the number of DEGs associated with each term. (<b>G</b>) Dot plot of KEGG pathway enrichment analysis for DEGs. Each dot represents a pathway, with the x-axis showing the enrichment score, dot color reflecting <span class="html-italic">p</span>-value significance, and dot size indicating the number of genes involved in each pathway.</p>
Full article ">Figure 2 Cont.
<p>Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of differentially expressed genes (DEGs). (<b>A</b>,<b>C</b>,<b>E</b>) Circular plots showing the top enriched GO terms for DEGs in the categories of biological process (BP) (<b>A</b>), cellular component (CC) (<b>C</b>), and molecular function (MF) (<b>E</b>). Each plot connects DEGs to their corresponding GO terms, with line colors reflecting the log fold change of the genes and circle sizes representing the number of associated genes for each term. (<b>B</b>,<b>D</b>,<b>F</b>) Dot plots summarizing the GO enrichment results for BP (<b>B</b>), CC (<b>D</b>), and MF (<b>F</b>). The x-axis indicates the enrichment score (−log10(<span class="html-italic">p</span>-value)), while dot colors represent <span class="html-italic">p</span>-value significance, and dot sizes correspond to the number of DEGs associated with each term. (<b>G</b>) Dot plot of KEGG pathway enrichment analysis for DEGs. Each dot represents a pathway, with the x-axis showing the enrichment score, dot color reflecting <span class="html-italic">p</span>-value significance, and dot size indicating the number of genes involved in each pathway.</p>
Full article ">Figure 2 Cont.
<p>Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of differentially expressed genes (DEGs). (<b>A</b>,<b>C</b>,<b>E</b>) Circular plots showing the top enriched GO terms for DEGs in the categories of biological process (BP) (<b>A</b>), cellular component (CC) (<b>C</b>), and molecular function (MF) (<b>E</b>). Each plot connects DEGs to their corresponding GO terms, with line colors reflecting the log fold change of the genes and circle sizes representing the number of associated genes for each term. (<b>B</b>,<b>D</b>,<b>F</b>) Dot plots summarizing the GO enrichment results for BP (<b>B</b>), CC (<b>D</b>), and MF (<b>F</b>). The x-axis indicates the enrichment score (−log10(<span class="html-italic">p</span>-value)), while dot colors represent <span class="html-italic">p</span>-value significance, and dot sizes correspond to the number of DEGs associated with each term. (<b>G</b>) Dot plot of KEGG pathway enrichment analysis for DEGs. Each dot represents a pathway, with the x-axis showing the enrichment score, dot color reflecting <span class="html-italic">p</span>-value significance, and dot size indicating the number of genes involved in each pathway.</p>
Full article ">Figure 2 Cont.
<p>Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of differentially expressed genes (DEGs). (<b>A</b>,<b>C</b>,<b>E</b>) Circular plots showing the top enriched GO terms for DEGs in the categories of biological process (BP) (<b>A</b>), cellular component (CC) (<b>C</b>), and molecular function (MF) (<b>E</b>). Each plot connects DEGs to their corresponding GO terms, with line colors reflecting the log fold change of the genes and circle sizes representing the number of associated genes for each term. (<b>B</b>,<b>D</b>,<b>F</b>) Dot plots summarizing the GO enrichment results for BP (<b>B</b>), CC (<b>D</b>), and MF (<b>F</b>). The x-axis indicates the enrichment score (−log10(<span class="html-italic">p</span>-value)), while dot colors represent <span class="html-italic">p</span>-value significance, and dot sizes correspond to the number of DEGs associated with each term. (<b>G</b>) Dot plot of KEGG pathway enrichment analysis for DEGs. Each dot represents a pathway, with the x-axis showing the enrichment score, dot color reflecting <span class="html-italic">p</span>-value significance, and dot size indicating the number of genes involved in each pathway.</p>
Full article ">Figure 3
<p>Gene set enrichment analysis (GSEA) and protein–protein interaction (PPI) network analysis. (<b>A</b>) GSEA plot of pathways enriched in the healthy group. The x-axis represents ranked genes, and the y-axis shows the enrichment score, highlighting pathways significantly upregulated in healthy individuals. (<b>B</b>) GSEA plot of pathways enriched in the diseased (IVDD) group. The plot illustrates pathways significantly upregulated in the diseased group, emphasizing key processes associated with disease progression. (<b>C</b>) PPI network of differentially expressed genes (DEGs). The network illustrates interactions among DEGs, with hub genes identified based on higher connectivity, indicating their potential regulatory roles in the associated pathways.</p>
Full article ">Figure 4
<p>Identification of disease characteristic genes by machine learning model. (<b>A</b>) Venn diagram showing the intersection of differentially expressed genes (DEGs) with glucose-related genes, yielding seven candidate genes for further analysis. (<b>B</b>) Boxplots comparing residuals between the random forest (RF) and support vector machine (SVM) models, where lower residuals indicate better performance of the RF model. (<b>C</b>) Reverse cumulative distribution of residuals in the RF and SVM models, further demonstrating the lower residuals achieved by the RF model. (<b>D</b>) Receiver operating characteristic (ROC) curves of the RF and SVM models. The RF model achieved an AUC of 1.000, compared to 0.931 for the SVM model, suggesting higher diagnostic precision. (<b>E</b>) Importance scores of the seven candidate genes calculated by the RF model. The red dashed line represents the error rate for the disease group (IVDD), the green dotted line represents the error rate for the healthy group, and the black solid line represents the overall error rate. As the number of trees increases, the overall error rate and individual class error rates stabilize, indicating model convergence. (<b>F</b>) Expression patterns of the three key genes (<span class="html-italic">Ins</span>, <span class="html-italic">Igfbp3</span>, and <span class="html-italic">Plod2</span>) identified by the RF model, selected for follow-up analyses due to their strong performance.</p>
Full article ">Figure 5
<p>Expression analysis and diagnostic performance of disease characteristic genes. (<b>A</b>) Heatmap displaying the expression levels of the three characteristic genes (<span class="html-italic">Ins</span>, <span class="html-italic">Igfbp3</span>, and <span class="html-italic">Plod2</span>) in diseased (neuropathic pain) and healthy groups. Red and blue indicate higher and lower expression levels, respectively. (<b>B</b>) Box plot showing the expression level of <span class="html-italic">Plod2</span>, which is significantly lower in the diseased group compared to the healthy group. (<b>C</b>) Box plot showing the expression level of <span class="html-italic">Igfbp3</span>, which is significantly higher in the diseased group compared to the healthy group. (<b>D</b>) Box plot showing the expression level of <span class="html-italic">INS</span>, which is significantly higher in the diseased group compared to the healthy group. Statistical significance for differential expression is presented in each figure. (<b>E</b>) Receiver operating characteristic (ROC) curve illustrating the diagnostic performance of the three genes (<span class="html-italic">Ins</span>, <span class="html-italic">Igfbp3</span>, and <span class="html-italic">Plod2</span>) in IVDD. The area under the curve (AUC) values demonstrates the predictive accuracy of these genes in distinguishing disease from healthy groups.</p>
Full article ">Figure 6
<p>Construction of a nomogram. (<b>A</b>) Nomogram developed based on the characteristic genes (<span class="html-italic">Ins</span>, <span class="html-italic">Igfbp</span>3, and <span class="html-italic">Plod2</span>) to predict the likelihood of the disease. Each gene is assigned a score, and the total score corresponds to a predicted probability of IVDD. (<b>B</b>) Clinical impact curve illustrating the negligible difference between the predicted and actual positive rates, validating the reliability of the nomogram for clinical application. (<b>C</b>) Decision curve analysis (DCA) demonstrating the net benefit of the nomogram across a range of threshold probabilities, indicating its high clinical value. (<b>D</b>) Calibration curve comparing the predicted positive rate from the nomogram with the actual observed positive rate. The curve’s alignment with the diagonal line indicates the nomogram’s strong diagnostic accuracy for IVDD.</p>
Full article ">Figure 7
<p>Immune infiltration analyses. (<b>A</b>) Heatmap displaying the relative percentages of 22 immune infiltrating cell types within the NP immune microenvironment. Each cell type’s proportion is calculated relative to the total immune cell population in individual samples, representing the composition of immune infiltration. (<b>B</b>) Correlation matrix showing the relationships between the 22 immune infiltrating cell types in the IVDD disease/NP group. Red and blue shades indicate positive and negative correlations, respectively. (<b>C</b>) Bar plot depicting the infiltration levels of immune cell types across individual samples from IVDD disease/NP patients, illustrating sample-specific immune variation. (<b>D</b>) Box plots comparing the levels of the 22 immune infiltrating cell types between diseased and healthy groups. Asterisks denote immune cell types that are differentially expressed between groups. (<b>E</b>) Correlation analysis between characteristic genes (<span class="html-italic">Igfbp3</span>, <span class="html-italic">Plod2</span>, and <span class="html-italic">Ins</span>) and the identified immune cell types. Positive correlations were observed between <span class="html-italic">Igfbp3</span>/<span class="html-italic">Plod2</span> and several immune cell types, while <span class="html-italic">Ins</span> expression was negatively associated with plasmacytoid dendritic cells and central memory CD8+ T cells.</p>
Full article ">Figure 7 Cont.
<p>Immune infiltration analyses. (<b>A</b>) Heatmap displaying the relative percentages of 22 immune infiltrating cell types within the NP immune microenvironment. Each cell type’s proportion is calculated relative to the total immune cell population in individual samples, representing the composition of immune infiltration. (<b>B</b>) Correlation matrix showing the relationships between the 22 immune infiltrating cell types in the IVDD disease/NP group. Red and blue shades indicate positive and negative correlations, respectively. (<b>C</b>) Bar plot depicting the infiltration levels of immune cell types across individual samples from IVDD disease/NP patients, illustrating sample-specific immune variation. (<b>D</b>) Box plots comparing the levels of the 22 immune infiltrating cell types between diseased and healthy groups. Asterisks denote immune cell types that are differentially expressed between groups. (<b>E</b>) Correlation analysis between characteristic genes (<span class="html-italic">Igfbp3</span>, <span class="html-italic">Plod2</span>, and <span class="html-italic">Ins</span>) and the identified immune cell types. Positive correlations were observed between <span class="html-italic">Igfbp3</span>/<span class="html-italic">Plod2</span> and several immune cell types, while <span class="html-italic">Ins</span> expression was negatively associated with plasmacytoid dendritic cells and central memory CD8+ T cells.</p>
Full article ">Figure 8
<p>Behavioral test PSNL-induced hypernasality on day 14 post-surgery. (<b>A</b>) Schematic illustration of behavioral test design and timeline. (<b>B</b>) Time course of mechanical allodynia, shown as the von Frey force threshold for withdrawal, in sham and PSNL groups. (** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001 vs. sham; sham, n = 6; PSNL, n = 6). Two-way repeated-measures analysis of variance (ANOVA) followed by Sidak’s multiple comparison. (<b>C</b>) Time course of the thermal withdrawal latency (s) in sham or PSNL mice. (**** <span class="html-italic">p</span> &lt; 0.0001 vs. sham; sham, n = 6; PSNL, n = 6). Two-way repeated-measures analysis of variance (ANOVA) followed by Sidak’s multiple comparison. (<b>D</b>) Representative track images and (<b>E</b>) Heatmap image of two-plate preference test at 30 °C versus 20 °C at 14 days post-operation. (<b>F</b>) Occupancy quantification of 30 °C versus 20 °C at baseline (left) and 14 days post-operation (right) (left: <span class="html-italic">p</span> = 0.5918, right: <span class="html-italic">p</span> = 0.6416; sham vs. PSNL; sham, n = 4; PSNL, n = 4). Unpaired <span class="html-italic">t</span>-test. (<b>G</b>) Changes in gait characteristics: toe spread of PSNL contralateral VS PSNL ipsilateral; (** <span class="html-italic">p</span> &lt; 0.01 vs. PSNL contralateral; PSNL, n = 3). Unpaired <span class="html-italic">t</span>-test. (<b>H</b>) Changes in gait characteristics: stride length (left) and stride width (right) of sham ipsilateral VS PSNL ipsilateral; (Left: <span class="html-italic">p</span> = 0.3717, right: <span class="html-italic">p</span> = 0.8295; sham vs. PSNL; sham, n = 4; PSNL, n = 4). Unpaired <span class="html-italic">t</span>-test. Unpaired <span class="html-italic">t</span>-test. (<b>I</b>) Changes in gait characteristics: toe spread of sham ipsilateral VS PSNL ipsilateral; (* <span class="html-italic">p</span> &lt; 0.05 vs. PSNL contralateral; PSNL, n = 3). Unpaired <span class="html-italic">t</span>-test.</p>
Full article ">Figure 9
<p>qRT-PCR showing mRNA expression levels of (<b>A</b>) <span class="html-italic">Ins1</span> (* <span class="html-italic">p</span> &lt; 0.05 vs. sham); (<b>B</b>) <span class="html-italic">Ins2</span> (<span class="html-italic">p</span> = 0.6788); (<b>C</b>) <span class="html-italic">Igfbp3</span> (* <span class="html-italic">p</span> &lt; 0.05 vs. sham); (<b>D</b>) <span class="html-italic">Plod2</span> (<span class="html-italic">p</span> = 0.6995) and (<b>E</b>) inflammatory related genes: <span class="html-italic">Il6</span> (** <span class="html-italic">p</span> &lt; 0.01 vs. sham), <span class="html-italic">Tnfα</span> (** <span class="html-italic">p</span> &lt; 0.01 vs. sham), and <span class="html-italic">Tgfβ</span> (<span class="html-italic">p</span> = 0.6836) in sham and PSNL at 14 days post-surgery. Gene expression was normalized to that of β-actin and is shown relative to sham, which is arbitrarily defined as 1 (sham vs. PSNL, n = 4~6/group). Unpaired <span class="html-italic">t</span>-test.</p>
Full article ">Figure 10
<p>Representative images of immunofluorescence of S100β in (<b>A</b>) sham and in (<b>B</b>) PSNL at 14 days post-surgery. Scale bars, 50 μm. Dashed rectangles indicate higher magnification (40×); scale bars, 20 μm. Arrows indicate S100β-positive cells. (<b>C</b>) Quantification of the numbers of sciatic nerve marked by S100β positive cells in sham and in PSNL (**** <span class="html-italic">p</span> &lt; 0.001 vs. sham; sham, n = 5; PSNL, n = 5). Unpaired <span class="html-italic">t</span>-test. Representative images of immunofluorescence of Cd86 and F4/80 in (<b>D</b>) sham and in (<b>E</b>) PSNL at 14 days post-surgery. Scale bars, 50 μm. Dashed rectangles indicate higher magnification (40×); scale bars, 20 μm. Arrows indicate Cd86 and F4/80 double-positive (co-staining) cells. (<b>F</b>) Quantification of the percentage of sciatic nerve marked by both Cd86 and F4/80 positive cells over total cells (Dapi) in sham and in PSNL (*** <span class="html-italic">p</span> &lt; 0.001 vs. sham; sham, n = 5; PSNL, n = 5). Unpaired <span class="html-italic">t</span>-test. Representative images of immunofluorescence of Cd11c. and F4/80 in (<b>G</b>) sham and in (<b>H</b>) PSNL at 14 days post-surgery. Scale bars, 50 μm. Dashed rectangles indicate higher magnification (40×); scale bars, 20 μm. Arrows indicate Cd11c negative and F4/80 positive cells. (<b>I</b>) Quantification of the percentage of sciatic nerve marked by Cd11c negative and F4/80 positive cells over total cells (Dapi) in sham and in PSNL (** <span class="html-italic">p</span> &lt; 0.01 vs. sham; sham, n = 5; PSNL, n = 5). Unpaired <span class="html-italic">t</span>-test.</p>
Full article ">Figure A1
<p>(<b>A</b>,<b>B</b>) Representative records from low-cost gait analysis, partially reflecting motor and sensory functions. Paw spread of the ipsilateral hind limb decreased following PSNL surgery. Blue indicates bilateral hind paws, while red represents forepaws. The upper section of the image shows the left fore and hind paw prints (left hind paw being the ipsilateral side) on an A4 paper, while the lower section shows the right fore and hind paw prints (right hind paw being the contralateral side). (<b>C</b>) Blood glucose levels measured at 0, 30, 60, and 90 min during the OGTT test using a glucometer. (<b>D</b>) Relative expression of the <span class="html-italic">Hif</span> gene measured by qPCR. (<b>E</b>,<b>F</b>) Representative images of the sciatic nerve including injury sites from the ipsilateral side under a 10× microscope. (<b>G</b>,<b>H</b>) Staining for CD16<sup>+</sup> cells in Sham and PSNL groups. White arrows indicate regions both DAPI and CD16 double positive. (<b>I</b>) Statistical analysis of CD16<sup>+</sup> cells in Sham and PSNL groups.</p>
Full article ">
11 pages, 5867 KiB  
Review
Prevention and Management of Recurrent Laryngeal Nerve Palsy in Minimally Invasive Esophagectomy: Current Status and Future Perspectives
by Yusuke Taniyama, Hiroshi Okamoto, Chiaki Sato, Yohei Ozawa, Hirotaka Ishida, Michiaki Unno and Takashi Kamei
J. Clin. Med. 2024, 13(24), 7611; https://doi.org/10.3390/jcm13247611 - 13 Dec 2024
Viewed by 401
Abstract
Recurrent laryngeal nerve palsy remains a significant complication following minimally invasive esophagectomy for esophageal cancer. Despite advancements in surgical techniques and lymphadenectomy precision, the incidence of recurrent laryngeal nerve palsy has not been improved. Recurrent laryngeal nerve palsy predominantly affects the left side [...] Read more.
Recurrent laryngeal nerve palsy remains a significant complication following minimally invasive esophagectomy for esophageal cancer. Despite advancements in surgical techniques and lymphadenectomy precision, the incidence of recurrent laryngeal nerve palsy has not been improved. Recurrent laryngeal nerve palsy predominantly affects the left side and may lead to unilateral or bilateral vocal cord paralysis, resulting in hoarseness, dysphagia, and an increased risk of aspiration pneumonia. While most cases of recurrent laryngeal nerve palsy are temporary and resolve within 6 to 12 months, some patients may experience permanent nerve dysfunction, severely impacting their quality of life. Prevention strategies, such as nerve integrity monitoring, robotic-assisted minimally invasive esophagectomy, and advanced dissection techniques, aim to minimize nerve injury, though their effectiveness varies. The management of recurrent laryngeal nerve palsy includes voice and swallowing rehabilitation, reinnervation techniques, and, in severe cases, surgical interventions such as thyroplasty and intracordal injection. As recurrent laryngeal nerve palsy can lead to significant postoperative respiratory complications, a multidisciplinary approach involving surgical precision, early detection, and comprehensive rehabilitation is crucial to improving patient outcomes and minimizing long-term morbidity in minimally invasive esophagectomy. This review article aims to inform esophageal surgeons and other clinicians about strategies for the prevention and management of recurrent laryngeal nerve palsy in esophagectomy. Full article
Show Figures

Figure 1

Figure 1
<p>Dissection of right RLN lymph nodes using a robot. (<b>a</b>) Dorsal side of lymph nodes along the right recurrent laryngeal nerve (RLN) are being dissected as the same plane with the dorsal side of the esophagus. This figure demonstrates that the lymphatic chain forming these nodes exhibits a mesenteric-like structure. (<b>b</b>) The lymphatic chain is dissected from the trachea, with the right recurrent laryngeal nerve and subclavian artery serving as the base, resembling a mesenteric structure. (<b>c</b>) As the lymphatic chain is dissected dorsally from the right subclavian artery, the recurrent laryngeal nerve (black arrowhead) naturally becomes visible under the thin membrane. (<b>d</b>) After dividing the esophageal branch of the recurrent laryngeal nerve and dissecting from the lateral wall of the trachea, en bloc resection of the lymphatic chain will be possible (yellow dots).</p>
Full article ">Figure 2
<p>Dissection of left RLN lymph nodes using a robot. (<b>a</b>) A stable surgical field is achieved by retracting the esophagus dorsally with gauze and using the robot to fix the trachea in place. (<b>b</b>) Dissection of the left side of the trachea from the lymphatic chain. The use of the robot allows for precise hemostasis while maneuvering over the trachea. (<b>c</b>) The sympathetic cardiac branch (black arrowhead) is revealed behind the thin membrane, as the lymphatic chain is flipped up. (<b>d</b>) The left RLN (white arrowhead) and its esophageal branch have been preserved, after flipping up the lymphatic chain.</p>
Full article ">Figure 3
<p>Schematic diagram of NIM (Nerve Integrity Monitoring). This diagram illustrates the mechanism of electrical stimulation of the vagus and RLN, which causes vocal cord movement detected via sensors attached to the endotracheal tube. The process is as follows: (<b>1</b>) The RLN is stimulated with a current of 0.5–1.0 mA. (<b>2</b>) The vocal cords move in response to the stimulation. (<b>3</b>) The sensor detects this vocal code movement. (<b>4</b>) The signal is transmitted as electrical impulses through the cord of the NIM endotracheal tube. (<b>5</b>) The stimulation is displayed as an electromyographic signal on the NIM monitor. Yellow arrows: Passage of electrical stimulation and signal to NIM system.</p>
Full article ">Figure 4
<p>Risk of RLN palsy associated with the application of strong forces during robotic esophagectomy. (<b>a</b>) Flexion of the left RLN (black dots) caused by the forceful elevation of the esophagus using the robot. (<b>b</b>) Flexion of the left RLN (black dots) resulting from powerful robotic dissection.</p>
Full article ">
9 pages, 210 KiB  
Article
The Acute Effect of Hot Water Immersion on Cardiac Function in Individuals with Cervical Spinal Cord Injury
by Ken Kouda, Motohiko Banno, Yasunori Umemoto, Tokio Kinoshita, Yukihide Nishimura, Yukio Mikami, Toshikazu Kubo and Fumihiro Tajima
J. Clin. Med. 2024, 13(24), 7593; https://doi.org/10.3390/jcm13247593 - 13 Dec 2024
Viewed by 403
Abstract
Background/Objectives: Thermotherapy is expected to assist in the prevention of arteriosclerosis and cardiovascular disease in individuals with spinal cord injuries. This study aimed to investigate the impact and underlying mechanisms of whole-body heat stress on cardiac function in patients with cervical spinal cord [...] Read more.
Background/Objectives: Thermotherapy is expected to assist in the prevention of arteriosclerosis and cardiovascular disease in individuals with spinal cord injuries. This study aimed to investigate the impact and underlying mechanisms of whole-body heat stress on cardiac function in patients with cervical spinal cord injury (CSCI) and healthy controls using head-out hot water immersion (HHWI). Methods: Eight male patients with complete motor CSCI and nine healthy controls were recruited. Participants were immersed for 60 min in water set at 2 °C above the resting esophageal temperature. Esophageal temperature, heart rate, and arterial pressure were monitored throughout the experiment. Before and after HHWI, echocardiography was used to measure indices of left ventricular diastolic capacity (E, E′, and A), left atrial contractility (A and A′), and left ventricular contractility [S′ and isovolumic acceleration (IVA)]. Results: Both groups exhibited an increase in body temperature and heart rate, while blood pressure remained stable. In the control group, there was a significant increase in E (67.0 ± 22.6 to 89.1 ± 13.6), E′ (9.5 ± 3.8 to 15.1 ± 4.1), A (50.0 ± 15.2 to 75.8 ± 18.2), A′ (8.1 ± 1.6 to 14.8 ± 5.9), S′ (8.7 ± 1.4 to 15.1 ± 4.5) and isovolumic acceleration (IVA) (104.2 ± 14.7 to 151.1 ± 20.6). In the CSCI group, only A (49.5 ± 9.9 to 56.9 ± 10.9) and IVA (94.4 ± 27.2 to 134.7 ± 27.7) showed a significant change. Conclusions: In the control group, heat stress increased left atrial contractility, left ventricular dilatation, and left ventricular contractility, while in patients with CSCI, left atrial contractility and left ventricular contractility improved, but there was no improvement in left ventricular diastolic function. This discrepancy in the impact of HHWI on cardiac function suggests that the sympathetic nervous system predominantly influences left ventricular dilatation during whole-body heat stress. However, other factors may also contribute to left atrial and ventricular contractility. Full article
(This article belongs to the Section Clinical Rehabilitation)
15 pages, 1995 KiB  
Systematic Review
The Comparative Efficacy of Burs Versus Piezoelectric Techniques in Third Molar Surgery: A Systematic Review Following the PRISMA Guidelines
by Rocco Franco, Mattia Di Girolamo, Carlo Franceschini, Sofia Rastelli, Mario Capogreco and Maurizio D’Amario
Medicina 2024, 60(12), 2049; https://doi.org/10.3390/medicina60122049 - 12 Dec 2024
Viewed by 515
Abstract
Background and Objectives: Third molar (wisdom tooth) extraction is one of the most common surgical procedures in oral and maxillofacial surgery. Traditional rotary instruments and burs have long been the standard tools for this procedure. However, recent advancements in surgical techniques, such [...] Read more.
Background and Objectives: Third molar (wisdom tooth) extraction is one of the most common surgical procedures in oral and maxillofacial surgery. Traditional rotary instruments and burs have long been the standard tools for this procedure. However, recent advancements in surgical techniques, such as piezoelectric surgery, have gained popularity due to their purported advantages in terms of precision, safety, and postoperative outcomes. This systematic review aims to evaluate the efficacy, safety, and clinical outcomes of third molar surgery performed using burs versus piezoelectric surgery. Materials and Methods: This systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A comprehensive literature search was performed using the PubMed, Scopus, Web of Science, and Cochrane databases to identify relevant studies published up until October 2024. Randomized controlled trials (RCTs), clinical trials, and comparative studies assessing third molar surgery using either burs or piezoelectric instruments were included. The primary outcomes evaluated were surgical time, postoperative pain, swelling, nerve damage, and healing time. The data extraction and quality assessment were performed independently by two reviewers using standardized tools, and any discrepancies were resolved by a third reviewer. Results: A total of five studies met the inclusion criteria, and the meta-analysis revealed that piezoelectric surgery resulted in significantly lower postoperative pain and swelling compared to traditional bur techniques (p < 0.05). Additionally, the incidence of nerve injury was lower in the piezoelectric group, though the difference was not statistically significant. Surgical time was found to be longer with piezoelectric devices, but this was offset by improved healing outcomes and patient comfort. Conclusions: Piezoelectric surgery offers a less traumatic alternative to traditional burs for third molar extraction, with reduced postoperative morbidity and enhanced patient outcomes. Although the longer surgical duration may be a drawback, the overall benefits, particularly in terms of pain management and tissue preservation, support the adoption of piezoelectric techniques in clinical practice. Further high-quality randomized trials are recommended to strengthen the evidence base for these findings. Full article
(This article belongs to the Special Issue Research on Oral and Maxillofacial Surgery)
Show Figures

Figure 1

Figure 1
<p>PRISMA flowchart.</p>
Full article ">Figure 2
<p>Forest plot evaluating VAS scores between two methods [<a href="#B26-medicina-60-02049" class="html-bibr">26</a>,<a href="#B27-medicina-60-02049" class="html-bibr">27</a>,<a href="#B28-medicina-60-02049" class="html-bibr">28</a>,<a href="#B29-medicina-60-02049" class="html-bibr">29</a>,<a href="#B30-medicina-60-02049" class="html-bibr">30</a>].</p>
Full article ">Figure 3
<p>Forest plot evaluating swelling between two methods [<a href="#B27-medicina-60-02049" class="html-bibr">27</a>,<a href="#B29-medicina-60-02049" class="html-bibr">29</a>].</p>
Full article ">Figure 4
<p>Bias assessment [<a href="#B26-medicina-60-02049" class="html-bibr">26</a>,<a href="#B27-medicina-60-02049" class="html-bibr">27</a>,<a href="#B28-medicina-60-02049" class="html-bibr">28</a>,<a href="#B29-medicina-60-02049" class="html-bibr">29</a>,<a href="#B30-medicina-60-02049" class="html-bibr">30</a>]. Green +: low risk of bias; red -: high rilsk of bias.</p>
Full article ">
Back to TopTop