[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

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,153)

Search Parameters:
Keywords = neural stem cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 5873 KiB  
Article
Injectable Tumoricidal Neural Stem Cell-Laden Hydrogel for Treatment of Glioblastoma Multiforme—An In Vivo Safety, Persistence, and Efficacy Study
by Jasmine L. King, Alain Valdivia, Shawn D. Hingtgen and S. Rahima Benhabbour
Pharmaceutics 2025, 17(1), 3; https://doi.org/10.3390/pharmaceutics17010003 - 24 Dec 2024
Abstract
Background/Objectives: Glioblastoma multiforme (GBM) is the most common high-grade primary brain cancer in adults. Despite efforts to advance treatment, GBM remains treatment resistant and inevitably progresses after first-line therapy. Induced neural stem cell (iNSC) therapy is a promising, personalized cell therapy approach that [...] Read more.
Background/Objectives: Glioblastoma multiforme (GBM) is the most common high-grade primary brain cancer in adults. Despite efforts to advance treatment, GBM remains treatment resistant and inevitably progresses after first-line therapy. Induced neural stem cell (iNSC) therapy is a promising, personalized cell therapy approach that has been explored to circumvent challenges associated with the current GBM treatment. Methods: Herein, we developed a chitosan-based (CS) injectable, biodegradable, in situ forming thermo-responsive hydrogel as a cell delivery vehicle for the treatment of GBM. Tumoricidal neural stem cells were encapsulated in the injectable CS hydrogel as stem cell therapy for treatment of post-surgical GBM. In this report, we investigated the safety of the injectable CS hydrogel in an immune-competent mouse model. Furthermore, we evaluated the persistence and efficacy of iNSC-laden CS hydrogels in a post-surgical GBM mouse model. Results: The injectable CS hydrogel was well tolerated in mice with no signs of chronic local inflammation. Induced neural stem cells (iNSCs) persisted in the CS hydrogels for over 196 days in comparison to 21 days for iNSCs (cell injection) only. GBM recurrence was significantly slower in mice treated with iNSC-laden CS hydrogels with a 50% increase in overall median survival in comparison to iNSCs (cell injection) only. Conclusions: Collectively, we demonstrated the ability to encapsulate, retain, and deliver iNSCs in an injectable CS hydrogel that is well tolerated with better survival rates than iNSCs alone. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the injectable CS hydrogel under physiological conditions.</p>
Full article ">Figure 2
<p>Schematic illustration depicting the transdifferentiation (TD) process of primary fibroblast to induced neural stem cells for implantation.</p>
Full article ">Figure 3
<p>Surgical procedure for in vivo persistence studies. (<b>1</b>) In step 1, a surgical incision was created to expose the intact skull, and an intracranial window (craniotomy) was created in the right hemisphere of the parietal skull plate using a microsurgical drill. (<b>2</b>) In step 2, using a surgical scope, an aspiration device was used to remove brain tissue to create a mock surgical resection cavity. (<b>3</b>) In step 3, a 5 µL of CS hydrogel solution containing 25,000 iNSCs was implanted into the resection cavity. The iNSC-CS hydrogel was given 1–2 min to settle before closing the wound with Vetbond tissue adhesive (3M 1469SB).</p>
Full article ">Figure 4
<p>In vivo retention and persistence post-implantation of iNSCs via direct injection or seeded in CS hydrogels. (<b>A</b>) In vivo persistence study design. (<b>B</b>) Representative BLI images collected up until signal loss for each group. (<b>C</b>,<b>D</b>) Summary graphs demonstrating the FLuc signal from iNSCs directly injected or encapsulated in CS hydrogel following delivery into the resection cavity. iNSC survival was represented as total FLuc signal from day 1 (* indicates <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA).</p>
Full article ">Figure 5
<p>Histology and safety of mice brain tissue following the resection and the CS hydrogel implantation. Histological images of brain tissue on days 3, 7, 30, 60, and 90 following the resection (n = 3/timepoint) and post-implantation of the CS hydrogel (n = 5/timepoint). (<b>A</b>,<b>B</b>) Resection control histological images at day 3 (<b>A</b>) and (<b>B</b>) a zoomed-in image of (<b>A</b>). (<b>C</b>,<b>D</b>) CS hydrogel histological images at day 3 (<b>C</b>) and (<b>D</b>) a zoomed-in image of (<b>C</b>). (<b>E</b>,<b>F</b>) Resection control histological images at day 7 (<b>E</b>) and (<b>F</b>) a zoomed-in image of (<b>E</b>). (<b>G,H</b>) CS hydrogel histological images at day 7 (<b>G</b>) and (<b>H</b>) a zoomed-in image of (<b>G</b>). (<b>I</b>,<b>J</b>) Resection control histological images at day 30 (<b>I</b>) and (<b>J</b>) a zoomed-in image of (<b>I</b>). (<b>K</b>,<b>L</b>) CS hydrogel histological images at day 30 (<b>K</b>) and (<b>L</b>) a zoomed-in image of (<b>K</b>). (<b>M</b>,<b>N</b>) Resection control histological images at day 60 (<b>M</b>) and (<b>N</b>) a zoomed-in image of (<b>M</b>). (<b>O</b>,<b>P</b>) CS hydrogel histological images at day 60 (<b>O</b>) and (<b>P</b>) a zoomed-in image of (<b>O</b>). (<b>Q</b>,<b>R</b>) Resection control histological images at day 90 (<b>Q</b>) and (<b>R</b>) a zoomed-in image of (<b>Q</b>). (<b>S</b>,<b>T</b>) CS hydrogel histological images at day 90 (<b>S</b>) and (<b>T</b>) a zoomed-in image of (<b>S</b>). R represents resection site, H represents hemorrhage (trauma-related), E represents edema (trauma-related), yellow circles represent swollen axons (trauma-related), and blue triangles represent pigment-laden macrophages. All scale bars represent 500 µm. All scale bars for zoomed-in images represent 50 µm.</p>
Full article ">Figure 6
<p>(<b>A</b>) Schematic illustration of tumor implantation, resection, and cell injection/implantation in vivo. (<b>B</b>) Fluorescent images of U87-MG mCh-FLuc tumors before and after resection and images of resection cavity using an Olympus MVX-10 microscope (1.6× magnification). White arrows represent positive tumor margins following resection.</p>
Full article ">Figure 7
<p>Delivery of tumoricidal iNSCs to inhibit progression of GBM in post-resection model. (<b>A</b>) Representative images of serial BLI showing tumor inhibition and regrowth in iNSC-sTR treated versus control-treated animals. (<b>B</b>) Summary graph depicting the tumor radiance of U87-MG FLuc overtime following post-resection treatment (* indicates <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005; *** <span class="html-italic">p</span> &lt; 0.001 by ANOVA). (<b>C</b>) Kaplan–Meier survival analysis demonstrating the survival of animals receiving iNSC-sTR therapy in comparison to control-treated animals (*** <span class="html-italic">p</span> &lt; 0.001 by log-rank test).</p>
Full article ">
11 pages, 4951 KiB  
Article
Stem Cells Within Three-Dimensional-Printed Scaffolds Facilitate Airway Mucosa and Bone Regeneration and Reconstruction of Maxillary Defects in Rabbits
by Mi Hyun Lim, Jung Ho Jeon, Sun Hwa Park, Byeong Gon Yun, Seok-Won Kim, Dong-Woo Cho, Jeong Hak Lee, Do Hyun Kim and Sung Won Kim
Medicina 2024, 60(12), 2111; https://doi.org/10.3390/medicina60122111 - 23 Dec 2024
Abstract
Background and Objectives: Current craniofacial reconstruction surgical methods have limitations because they involve facial deformation. The craniofacial region includes many areas where the mucosa, exposed to air, is closely adjacent to bone, with the maxilla being a prominent example of this structure. [...] Read more.
Background and Objectives: Current craniofacial reconstruction surgical methods have limitations because they involve facial deformation. The craniofacial region includes many areas where the mucosa, exposed to air, is closely adjacent to bone, with the maxilla being a prominent example of this structure. Therefore, this study explored whether human neural-crest-derived stem cells (hNTSCs) aid bone and airway mucosal regeneration during craniofacial reconstruction using a rabbit model. Materials and Methods: hNTSCs were induced to differentiate into either mucosal epithelial or osteogenic cells in vitro. hNTSCs were seeded into polycaprolactone scaffold (three-dimensionally printed) that were implanted into rabbits with maxillary defects. Four weeks later, tissue regeneration was analyzed via histological evaluation and immunofluorescence staining. Results: In vitro, hNTSCs differentiated into both mucosal epithelial and osteogenic cells. hNTSC differentiation into respiratory epithelial cells was confirmed by Alcian Blue staining, cilia in SEM, and increased expression levels of FOXJ1 and E-cadherin through quantitative RT-PCR. hNTSC differentiation into bone was confirmed by Alizarin Red staining, increased mRNA expression levels of BMP2 (6.1-fold) and RUNX2 (2.3-fold) in the hNTSC group compared to the control. Four weeks post-transplantation, the rabbit maxilla was harvested, and H&E, SEM, and immunohistofluorescence staining were performed. H&E staining and SEM showed that new tissue and cilia around the maxillary defect were more prominent in the hNTSC group. Also, the hNTSCs group showed positive immunohistofluorescence staining for acetylated α-tubulin and cytokerin-5 compared to the control group. Conclusions: hNTSCs combined with PCL scaffold enhanced the regeneration of mucosal tissue and bone in vitro and promoted mucosal tissue regeneration in the in vivo rabbit model. Full article
(This article belongs to the Special Issue New Insights into Plastic and Reconstructive Surgery)
Show Figures

Figure 1

Figure 1
<p>Differentiation of hNTSCs into epithelial cells. (<b>A</b>) Alcian Blue staining revealing mucus production. Scale bars: 100 µm (<b>B</b>) SEM of cilia. Scale bars: 1.0 µm. (<b>C</b>) Real-time PCR data derived on days 0 and 45 after epithelial differentiation were induced. Bars: Relative expression levels (±SDs). * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Cell seeding into artificial maxillary grafts and pre-implantation culture. (<b>A</b>) hNTSC seeding into AMG in a spinner flask. hNTSC sheets covered the AMG after 3 days of culture in osteogenic induction medium before implantation of AMG-hNTSCs into rabbits. (<b>B</b>) An optical microscopic image of AMG seeded with hNTSCs after 3 days of culture (upper panel) and a confocal microscope image (with z-stack projections) after staining for F-actin (red). The nuclei were stained with DAPI (blue). Scale bars: 200 and 100 µm, respectively. (<b>C</b>) Images of CTRL (non-induced A-hNTSCs) and A-hNTSCs stained with Alizarin Red S, a dye used to detect calcium deposition, after 21 days of incubation in osteogenic differentiation medium. (<b>D</b>) The mineralization was quantified by extraction of Alizarin Red S dye using the CPC extraction method, and absorbance was measured at 570 nm. * <span class="html-italic">p</span> &lt; 0.05 compared with CTRL group. (<b>E</b>) The expression levels of BMP2 and RUNX2 after 21 days of incubation in osteogenic differentiation medium as revealed by real-time PCR. Bars: Relative expression levels (±SDs). ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Surgical and sacrifice procedures and evaluation of ciliary regeneration. (<b>A</b>) Images taken during surgery (<b>top</b>) and sacrifice (<b>bottom</b>). (<b>B</b>) H&amp;E staining of paraffin-embedded sections (scale bars: 1000 µm and 100 µm, respectively), and (<b>C</b>) SEM images obtained at 4 weeks after implantation of AMG or A-hNTSCs. Scale bars: 10 µm.</p>
Full article ">Figure 4
<p>Immunohistofluorescence staining of hNTSCs in maxillary defects after implantation of AMG or A-hNTSCs. Confocal microscopy images (z-stack projections) of AMG and A-hNTSCs after staining of paraffin-embedded sections (a) with an antibody against (<b>A</b>) HuNu and (<b>B</b>) double-staining with antibodies against acetylated α-tubulin (green) and cytokeratin-5 (red) at 4 weeks. The nuclei were labeled with DAPI (blue). Scale bars: (<b>A</b>,<b>B</b>): upper panels 50 µm; lower panels 20 µm.</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 313
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

Figure 1

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 ">
23 pages, 6263 KiB  
Article
Development of a Herb-Based Dietary Ingredient with Potential Nootropic Properties: From Bench to Bedside
by Pau Navarro, Justyna Meissner, José Luis Mullor, Nuria Caturla and Jonathan Jones
Appl. Sci. 2024, 14(24), 11869; https://doi.org/10.3390/app142411869 - 19 Dec 2024
Viewed by 362
Abstract
Cognitive decline is a natural process that occurs during aging. Several natural solutions called nootropics exist that can help mitigate this process. However, in some cases, there is a lack of scientific evidence on their potential mechanisms and efficacy. To this end, a [...] Read more.
Cognitive decline is a natural process that occurs during aging. Several natural solutions called nootropics exist that can help mitigate this process. However, in some cases, there is a lack of scientific evidence on their potential mechanisms and efficacy. To this end, a total of nine different herb extracts with potential nootropic activity were evaluated. These herb extracts were tested in human neural stem cell cultures exposed to oxidative stress, where reactive oxygen/nitrogen species, cell survival and trophic factor expression were analyzed. Of these extracts, Salvia officinalis (sage) was observed to possess the most significant effect. Its combination with rutin demonstrated additional benefits than with only sage. A randomized, double-blind placebo-controlled clinical study was then performed, testing the effects of the combination at two different doses for 12 weeks in healthy volunteers aged 50+ with mild cognitive impairment. As a result, the blend significantly improved several characteristics related to memory, particularly memory recall, with a more noticeable effect in the higher dose. Therefore, this study reveals the potential nootropic effect of a blend comprising Salvia officinalis and rutin. This blend could be a candidate dietary ingredient with potential nootropic properties. Full article
(This article belongs to the Special Issue Recent Applications of Plant Extracts in the Food Industry)
Show Figures

Figure 1

Figure 1
<p>MTT analysis in neural stem cells and in presence of the botanical extracts (bilberry, blueberry, bacopa, sage, saffron, arctic root, ginkgo, ginseng, gotu kola), using DMSO as control. Red dash base level of control with DMSO. * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>ROS analysis of botanical extracts in neural stem cells exposed to H<sub>2</sub>O<sub>2</sub> and in the presence of the botanical extracts (bilberry, blueberry, bacopa, sage, saffron, arctic root, ginkgo, ginseng, gotu kola). Dash line indicates control with H<sub>2</sub>O<sub>2</sub> levels. * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>RNS analysis in neural stem cells exposed to H<sub>2</sub>O<sub>2</sub> and in the presence of the botanical extracts (bilberry, blueberry, bacopa, sage, saffron, arctic root, ginkgo, ginseng, gotu kola). Dashed line indicates control with H<sub>2</sub>O<sub>2</sub> levels. * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Caspase-9 gene expression analysis in neural stem cells exposed to H<sub>2</sub>O<sub>2</sub> and in the presence of the botanical extracts. Dashed line indicates control with H<sub>2</sub>O<sub>2</sub> levels. * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001. Ratio corresponds to the gene relative expression ratio.</p>
Full article ">Figure 5
<p>Expression levels of trophic factors NRF2, NGF and VEGF in neural stem cells exposed to H<sub>2</sub>O<sub>2</sub> and in the presence of the botanical extracts. Dashed line indicates control with H<sub>2</sub>O<sub>2</sub> levels. * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001. Ratio corresponds to the gene relative expression ratio. NRF2: nuclear factor erythroid 2-related factor 2, NGF: nerve growth factor, VEGF: vascular endothelial growth factor.</p>
Full article ">Figure 5 Cont.
<p>Expression levels of trophic factors NRF2, NGF and VEGF in neural stem cells exposed to H<sub>2</sub>O<sub>2</sub> and in the presence of the botanical extracts. Dashed line indicates control with H<sub>2</sub>O<sub>2</sub> levels. * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001. Ratio corresponds to the gene relative expression ratio. NRF2: nuclear factor erythroid 2-related factor 2, NGF: nerve growth factor, VEGF: vascular endothelial growth factor.</p>
Full article ">Figure 6
<p>Expression of BDNF in NSC cultured in the presence of sage or sage/rutin extract. Dashed line indicates control levels. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>AChE activity in hippocampal cell cultures in the presence of sage/rutin at different concentrations. Dashed line indicates control with H<sub>2</sub>O<sub>2</sub> levels. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 8
<p>MoCA general score of the healthy volunteers. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>ADAS-Cog Scores. ** <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; 0.05 vs. placebo.</p>
Full article ">Figure 10
<p>ADAS-Cog subscores. † <span class="html-italic">p</span> &lt; 0.05 vs. placebo.</p>
Full article ">
15 pages, 1213 KiB  
Review
Mesenchymal Stem Cells and Their Extracellular Vesicles: Therapeutic Mechanisms for Blood–Spinal Cord Barrier Repair Following Spinal Cord Injury
by Masahito Nakazaki, Takahiro Yokoyama, Karen L. Lankford, Ryosuke Hirota, Jeffery D. Kocsis and Osamu Honmou
Int. J. Mol. Sci. 2024, 25(24), 13460; https://doi.org/10.3390/ijms252413460 - 16 Dec 2024
Viewed by 424
Abstract
Spinal cord injury (SCI) disrupts the blood–spinal cord barrier (BSCB) exacerbating damage by allowing harmful substances and immune cells to infiltrate spinal neural tissues from the vasculature. This leads to inflammation, oxidative stress, and impaired axonal regeneration. The BSCB, essential for maintaining spinal [...] Read more.
Spinal cord injury (SCI) disrupts the blood–spinal cord barrier (BSCB) exacerbating damage by allowing harmful substances and immune cells to infiltrate spinal neural tissues from the vasculature. This leads to inflammation, oxidative stress, and impaired axonal regeneration. The BSCB, essential for maintaining spinal cord homeostasis, is structurally similar to the blood–brain barrier. Its restoration is a key therapeutic target for improving outcomes in SCI. Mesenchymal stromal/stem cells (MSCs) and their secreted extracellular vesicles (MSC-EVs) have gained attention for their regenerative, immunomodulatory, and anti-inflammatory properties in promoting BSCB repair. MSCs enhance BSCB integrity by improving endothelial–pericyte association, restoring tight junction proteins, and reducing inflammation. MSC-EVs, which deliver bioactive molecules, replicate many of MSCs’ therapeutic effects, and offer a promising cell-free alternative. Preclinical studies have shown that both MSCs and MSC-EVs can reduce BSCB permeability, promote vascular stability, and support functional recovery. While MSC therapy is advancing in clinical trials, MSC-EV therapies require further optimization in terms of production, dosing, and delivery protocols. Despite these challenges, both therapeutic approaches represent significant potential for treating SCI by targeting BSCB repair and improving patient outcomes. Full article
(This article belongs to the Special Issue Activation of the Blood–Brain Barrier and Neurological Dysfunction)
Show Figures

Figure 1

Figure 1
<p>Blood–Spinal Cord Barrier Disruption Following Spinal Cord Injury (SCI).(<b>A</b>) Schematic illustration depicting the cellular components of the blood–spinal cord barrier (BSCB) under normal conditions, with astrocytes, pericytes, and endothelial cells maintaining BSCB integrity. Following SCI, the barrier is disrupted, allowing infiltration of harmful substances such as pathogens, pro-inflammatory cells, and neurotoxic molecules into the spinal cord parenchyma. The area marked with an asterisk (*) in panel (<b>A</b>) is shown in an enlarged view in panel (<b>B</b>). (<b>B</b>) Tight junction breakdown is shown, where key proteins such as N-cadherin, occludin, and ZO-1 dissociate, resulting in increased BSCB permeability. This disruption leads to a loss of selective permeability, impaired nutrient and signaling molecule exchange, enlargement of blood vessels, neuroinflammation, and secondary injury processes that exacerbate neurotoxicity.</p>
Full article ">Figure 2
<p>MSC and MSC-EV Mediated Repair of the Blood–Spinal Cord Barrier Post-SCI. (<b>A</b>). Intravenous (I.V) infusion of mesenchymal stem cells (MSCs) or their extracellular vesicles (MSC-EVs) plays a pivotal role in repairing the blood–spinal cord barrier (BSCB) following spinal cord injury (SCI). MSC-EVs specifically target M2 macrophages, enhancing their anti-inflammatory and reparative functions. This leads to the secretion of bioactive molecules that aid in endothelial cell stabilization and reduce inflammation at the injury site. The area marked with an asterisk (*) in panel (<b>A</b>) is shown in an enlarged view in panel (<b>B</b>). (<b>B</b>). MSC-EVs promote the restoration of the BSCB by upregulating tight junction proteins, such as occludin, ZO-1, and N-cadherin. These proteins help re-establish tight junction integrity, improving vascular stabilization and reducing the infiltration of harmful molecules into the spinal cord. Anti-inflammatory cytokines like TGF-β and IL-10, secreted by M2 macrophages, further support long-term BSCB repair and functional recovery.</p>
Full article ">
13 pages, 2884 KiB  
Article
A High-Throughput Neurosphere-Based Colony Formation Assay to Test Drug and Radiation Sensitivity of Different Patient-Derived Glioblastoma Lines
by Manoj Kumar, Lauren C. Nassour-Caswell, Hasan Alrefai, Joshua C. Anderson, Taylor L. Schanel, Patricia H. Hicks, Rex Cardan and Christopher D. Willey
Cells 2024, 13(23), 1995; https://doi.org/10.3390/cells13231995 - 3 Dec 2024
Viewed by 606
Abstract
The gold standard assay for radiation response is the clonogenic assay, a normalized colony formation assay (CFA) that can capture a broad range of radiation-induced cell death mechanisms. Traditionally, this assay relies on two-dimensional (2D) cell culture conditions with colonies counted by fixing [...] Read more.
The gold standard assay for radiation response is the clonogenic assay, a normalized colony formation assay (CFA) that can capture a broad range of radiation-induced cell death mechanisms. Traditionally, this assay relies on two-dimensional (2D) cell culture conditions with colonies counted by fixing and staining protocols. While some groups have converted these to three-dimensional (3D) conditions, these models still utilize 2D-like media compositions containing serum that are incompatible with stem-like cell models such as brain tumor initiating cells (BTICs) that form self-aggregating spheroids in neural stem cell media. BTICs are the preferred patient-derived model system for studying glioblastoma (GBM) as they tend to better retain molecular and phenotypic characteristics of the original tumor tissue. As such, it is important that preclinical radiation studies should be adapted to BTIC conditions. In this study, we describe a series of experimental approaches for performing CFA experiments with BTIC cultures. Our results indicate that serum-free clonogenic assays are feasible for combination drug and radiation testing and may better facilitate translatability of preclinical findings. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) 39P-RT cells were seeded in the inner 60 wells of a 96-well plate at different indicated cell numbers and treated with DMSO (vehicle) or Drug A and after 2 h were irradiated with different doses of radiation. After 12 days of colony formation, colonies were stained with 0.2 μg/mL of Calcein AM (green) and imaged at 4× magnification on the Cytation 5, using 120 μm as the cutoff value for colony counting. Colony numbers are indicated in white in each well. (<b>B</b>) The semi-log clonogenic curve was plotted with standard error bars. Only the 2 Gy dose was significantly different between Drug A and vehicle (<span class="html-italic">p</span> &lt; 0.01) after analyzing two-way ANOVA. (<b>C</b>) 14P mCherry neurospheres suspension was used to seed a clear bottom 96-well black-wall plate, and after two days of incubation in a CO<sub>2</sub> incubator at 37 °C, these neurospheres were fixed with 0, 0.25, 0.47, or 0.84% PFA and then stored at 4 °C. Serial imaging was carried out using the Cytation 5 Imager from 2 h to 13 days using the 590 nm Filter/LED cube (PM 1225002), which has an excitation of 586/15 nm and an emission of 647/57 nm. Created in BioRender.com.</p>
Full article ">Figure 2
<p>(<b>A</b>) 14P NLS mCherry cells were seeded in the inner 60 wells of two 96-well plates. After 24 h, cells were treated with drug B, drug C, and their combination. After 2 h of drug treatment, one plate was irradiated with 4 Gy while the other was sham irradiated. After 12 days of colony formation, colonies were stained with 0.2 μg/mL of Calcein AM (green) and imaged at 4× magnification on the Cytation 5. Colony counts were made using a 100 μm size cutoff. (<b>B</b>) Surviving fraction was calculated for each drug/drug combination condition followed by normalization with the 4 Gy/0 μM condition (ordinary one-way ANOVA statistics are shown with SEM). (<b>C</b>) Relative colony formations are plotted with normalization only to the 0 Gy/0 μM condition (two-way ANOVA is used for statistical analysis). For (<b>B</b>,<b>C</b>), * indicates <span class="html-italic">p</span> value &lt; 0.05; ** indicates <span class="html-italic">p</span> &lt; 0.01; **** indicates <span class="html-italic">p</span> &lt; 0.0001; ‘ns’ stands for non-significant. (<b>D</b>) 14P BTIC cells were plated at 50 cells/well in 96-well plates and allowed to grow as neurospheres for 5 days. After 5 days, neurospheres were treated with drug X, and after 5 h, they were irradiated with sham (0 Gy) or 5 Gy of radiation. After 12 days of colony/neurosphere formation, colonies were stained with 0.2 μg/mL of Calcein AM (green) and imaged at 4× magnification on Cytation 5. (<b>E</b>) Relative change in colony sizes is plotted and two-way ANOVA is used for statistical analysis. * indicates <span class="html-italic">p</span> value &lt; 0.05; **** indicates <span class="html-italic">p</span> &lt; 0.0001; ‘ns’ stands for non-significant.</p>
Full article ">Figure 3
<p>(<b>A</b>,<b>C</b>) J14P and J14P-RT mCherry, 300 cells/well, were seeded in 96-well plate, and after 24 h, were irradiated with different doses of radiations (0, 2, 4, 6, and 8 Gy). After 13 days of colony/neurosphere formation, colonies were fixed with 0.84% PFA, stained with 4 μg/mL of WGA Alexa Fluor 488 (green), and imaged at 4× magnification using both green and red channels on the Cytation 5. Colonies of size 120 μm or above were counted as true colonies. (<b>B</b>,<b>D</b>) Relative colony formations are plotted and two-way ANOVA is used for statistical analysis. ** indicates <span class="html-italic">p</span> &lt; 0.01; *** indicates <span class="html-italic">p</span> &lt; 0.001; ‘ns’ stands for non-significant.</p>
Full article ">Figure 4
<p>(<b>A</b>) 14P mCherry and 14P-RT mCherry neurospheres were plated at 300 cells/well in 96-well plates and irradiated with different radiation doses in a fractionated approach (4 days between fractions). Following fixation with 0.84% PFA, cells were stained with 4 μg/mL of WGA Alexa Fluor 488 (green). (<b>B</b>) Relative colony formations are plotted and two-way ANOVA is used for statistical analysis. ** indicates <span class="html-italic">p</span> value &lt; 0.01 and ‘ns’ stands for non-significant.</p>
Full article ">
26 pages, 4558 KiB  
Article
A Comprehensive Functional Investigation of the Human Translocator Protein 18 kDa (TSPO) in a Novel Human Neuronal Cell Knockout Model
by Stefanie Bader, Tatjana Jahner, Anett Dörfelt, Doris Melchner, Iseline Cardon, Heiko I. Siegmund, Christoph Brochhausen, Rainer Rupprecht, Vladimir M. Milenkovic and Christian H. Wetzel
Int. J. Mol. Sci. 2024, 25(23), 12882; https://doi.org/10.3390/ijms252312882 - 29 Nov 2024
Viewed by 508
Abstract
The translocator protein 18 kDa (TSPO) is a multifunctional outer mitochondrial membrane protein associated with various aspects of mitochondrial physiology and multiple roles in health and disease. Here, we aimed to analyse the role of TSPO in the regulation of mitochondrial and cellular [...] Read more.
The translocator protein 18 kDa (TSPO) is a multifunctional outer mitochondrial membrane protein associated with various aspects of mitochondrial physiology and multiple roles in health and disease. Here, we aimed to analyse the role of TSPO in the regulation of mitochondrial and cellular functions in a human neuronal cell model. We used the CRISPR/Cas9 technology and generated TSPO knockout (KO) and control (CTRL) variants of human-induced pluripotent stem cells (hiPSCs). In a multimodal phenotyping approach, we investigated cellular and mitochondrial functions in neural progenitor cells (NPCs), astrocytes, and neurons differentiated from hiPSC CTRL and TSPO KO cell lines. Our analysis revealed reduced mitochondrial respiration and glycolysis, altered Ca2+ levels in the cytosol and mitochondrial matrix, a depolarised MMP, and increased levels of reactive oxygen species, as well as a reduced cell size. Notably, TSPO deficiency was accompanied by reduced expression of the voltage-dependent anion channel (VDAC). We also observed a reduced TSPO and VDAC expression in cells derived from patients suffering from major depressive disorder (MDD). Considering the modulatory function of TSPO and the similar functional phenotype of cells derived from patients with depression, we discuss a role of TSPO in the etiology or pathology of MDD. In summary, our findings indicate a general impairment of mitochondrial function in TSPO knockout (KO) cells. This deepens our insight into the intricate role of TSPO in a range of physiological and pathological processes. Full article
(This article belongs to the Special Issue Mitochondria in Human Health and Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Characterisation of TSPO knockout model. (<b>A</b>) Translocator protein 18 kDa (TSPO) gene deletion in human-induced pluripotent stem cells. Schematic overview displaying the CRISPR/Cas9 sgRNA #116 and #126 targeting sites on exon 2 of the TSPO gene, along with their respective PAM sequences (cyan). Asterisk * represents end of translation (stop codon). Successful TSPO knockout in both alleles was confirmed using DNA sequencing. (<b>B</b>) Western blot analysis of TSPO protein expression in hiPSC control (CTRL1, CTRL2, and CTRL3) and knockout (KO1, KO2, and KO3) lines, demonstrating a complete loss of TSPO. BTUB1 was used as a loading control. (<b>C</b>) Immunofluorescence staining demonstrates the presence of mature astrocyte markers, including GFAP, ALDH1L1, S100β, and EAAT1. Scale bar represents 20 µm. Representative examples of (<b>D</b>) spontaneous and (<b>E</b>) ATP-responsive Ca<sup>2+</sup> transients in astrocytes loaded with Fura-2/AM. In (<b>E</b>), the gliotransmitter ATP was added at a final concentration of 100 µM. Representative Fura-2 ratio images display brighter cells post-ATP treatment, indicating increased intracellular calcium levels (left). (<b>F</b>–<b>H</b>) Characterisation of hiPSC-derived neurons. (<b>F</b>) Immunofluorescence staining at day 21 of differentiation reveals the expression of neuronal markers in the induced neurons. The neurons display typical neuronal cytoskeleton proteins MAP-2 and Tuj-1, as well as the presynaptic synaptophysin (SYN) and post-mitotic neuronal nuclear marker NeuN. Immunostaining for VGLUT1 and postsynaptic PSD95 point to glutamatergic synapses. Scale bar indicates 20 µM. (<b>G</b>) Electron micrographs provide high-resolution visualisation of neuronal morphology after 21 days of differentiation. (<b>H</b>) Changes and oscillations in intracellular Ca<sup>2+</sup> concentrations visualised via Fura-2/AM live-cell imaging demonstrate the spontaneous activity of induced neurons at day 21 of differentiation. (<b>I</b>) First signs of a synchronous activity pattern assessed with the HD-MEA system from Maxwell technologies at day 30 of differentiation. (<b>J</b>) TSPO knockout in hiPSC-derived neural progenitors, astrocytes, and neurons. TSPO gene deletion was further confirmed by TSPO antibody co-staining, using VDAC1 as a mitochondrial marker. Scale bar indicates 20 µm. Control of TSPO immunofluorescence (without 1st antibody) is shown in <a href="#app1-ijms-25-12882" class="html-app">Supplementary Figure S2</a>.</p>
Full article ">Figure 2
<p>Pregnenolone synthesis and mRNA expression of steroidogenic enzymes. (<b>A</b>) TSPO knockout resulted in significantly lower basal pregnenolone production in hiPSC-derived astrocytes compared to their TSPO-expressing CTRL cells. Dot blots represent normalised concentrations ± SEM using independent samples <span class="html-italic">t</span>-test. (<b>B</b>) Relative mRNA levels of key steroidogenic proteins (CYP11A1, CYP17A1, HSD3B1, and StAR) in hiPSC-derived astrocytes (control) and the adrenocarcinoma cell line H295-R related to the very low gene expression in C20 microglial cells. Bar graphs represent the mean fold change ± SEM. Asterisks in the figure represent ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Mitochondrial respiration in TSPO knockout and control cells. The oxygen consumption rate (OCR) was measured in TSPO KO and CTRL cells to assess the basal conditions and the effects of sequential injections of oligomycin (Oligo), carbonyl cyanide-4-(trifluoromethoxy)-phenylhydrazone (FCCP), and rotenone/antimycin A (Rot/AA) targeting different complexes of the electron transport chain. Representative examples of kinetic profiles of the Agilent Seahorse XF Mito Stress Test are shown for (<b>A</b>) NPCs and (<b>D</b>) astrocytes. Key respiratory parameters, including basal OCR, maximal respiration, proton leak, ATP-related oxygen consumption, spare respiratory capacity, and non-mitochondrial respiration, were compared between the KO and CTRL groups in (<b>B</b>) NPCs and (<b>E</b>) astrocytes. Total ATP content was quantified in (<b>C</b>) NPCs and (<b>F</b>) astrocytes using a bioluminescence assay. Dot plots display the normalised mean OCR values for NPCs and astrocytes. ATP levels in nM normalised to the total protein amount [µg/mL] are shown for the ATP content in NPCs and astrocytes. Data are presented as mean ± SEM using independent samples <span class="html-italic">t</span>-test. (<b>G</b>–<b>I</b>) Effects of TSPO gene deletion on mitochondrial membrane potential. TSPO deficiency resulted in a significant reduction in mitochondrial membrane potential in knockout cells of hiPSC-derived neural progenitors (<b>G</b>), astrocytes (<b>H</b>), and neurons (<b>I</b>) (<span class="html-italic">p</span> &lt; 0.0001, Mann–Whitney U test). The MMP in NPCs and neurons is indicated by the JC-1 fluorescence ratio of red/green (JC-1 aggregates/monomers) and in astrocytes by the ratio of TMRE/MTG fluorescence. Dot plots show mean red/green or TMRE/MTG ratio ± SEM of <span class="html-italic">n</span> = 3 biological replicates of several individual cell lines (NPC: CTRL <span class="html-italic">n</span> = 3, KO <span class="html-italic">n</span> = 3; Astro: CTRL <span class="html-italic">n</span> = 2, KO <span class="html-italic">n</span> = 2; Neuron: CTRL <span class="html-italic">n</span> = 2, KO <span class="html-italic">n</span> = 2). Representative fluorescence microscopy images of NPCs and neurons loaded with the cationic dye JC-1 and of astrocytes loaded with the cationic dye TMRE and the mitochondria-specific dye MitoTracker Green. Scale bar indicates 20 µm. (<b>J</b>) Impact of TSPO gene deletion on cytosolic and mitochondrial Ca<sup>2+</sup>. TSPO knockout cells showed significantly reduced cytosolic Ca<sup>2+</sup> levels, as indicated by the Fura-2 fluorescence ratio F<sub>340</sub>/<sub>380</sub>, while the mitochondrial Ca<sup>2+</sup> concentration was significantly increased in TSPO KO NPCs, astrocytes, and neurons, as indicated by Rhod-2 mean fluorescence intensity (MFI) ± SEM. Exact statistical values are given in <a href="#app1-ijms-25-12882" class="html-app">Supplementary Table S4</a>. Asterisks in the figure represent <span class="html-italic">p</span>-values as follows: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Effect of TSPO deficiency on VDAC1 gene expression and protein stability. (<b>A</b>) Deletion of <span class="html-italic">TSPO</span> gene led to decreased expression of VDAC1 protein in NPCs and astrocytes. Representative Western blots depict the expression of the housekeeper gene BTUB1 at 55 kDa and VDAC1 at 32 kDa in NPCs and astrocytes. Bar graphs show densitometric, relative mean of VDAC expression. Individual values were normalised to the respective expression level of BTUB1 and related to CTRL mean. Data are presented as mean ± SEM using Mann–Whitney U test. (<b>B</b>) qRT-PCR analysis revealed no significant differences in mRNA transcript levels of VDAC1 in TSPO knockout NPCs compared to CTRL cells. Bar graphs show fold change ± SEM related to CTRL of <span class="html-italic">n</span> = 2 biological replicates. (<b>C</b>) CTRL and KO cells were treated with cycloheximide (100 µg/mL) for the indicated times, and endogenous VDAC1 was detected using a specific anti-VDAC1 antibody. Values were normalised to the expression level of BTUB1 and were related to the initial VDAC1 protein level (0 h of CHX) for each group. Data are shown as relative mean ± SEM of <span class="html-italic">n</span> = 9 biological replicates. Asterisks in the figure represent **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Glycolytic rates in TSPO knockout and control cells. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured in TSPO KO and CTRL cells. Following basal measurement of OCR and ECAR to determine the basal proton efflux rate (PER), rotenone and antimycin A (Rot/AA) were added to determine compensatory glycolysis and basal glycolysis (glycoPER) by blocking mitochondrial respiration. Subsequently, 2-deoxy-D-glucose (2-DG), an inhibitor of glycolysis by competitively binding hexokinase, was injected to ensure that the observed PER was caused by glycolysis. Representative kinetic profiles of the Agilent Seahorse Glycolytic Rate assay and key glycolytic parameters are shown for (<b>A</b>) neural progenitors (NPCs) and (<b>B</b>) astrocytes. Key glycolytic parameters, including basal glycolysis, basal PER, compensatory glycolysis, post-2DG acidification, and mitoOCR/glycoPER, were compared between KO and CTRL groups. Dot plots display normalised mean PER values ± SEM using independent samples <span class="html-italic">t</span>-test or Mann–Whitney U test for NPCs and astrocytes. (<b>C</b>,<b>D</b>) ATP production rates in TSPO knockout and control cells. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured, and ATP production rates were calculated for TSPO KO and CTRL cells after sequential addition of the metabolic modulators oligomycin (oligo) and rotenone/antimycin A (Rot/AA). Representative kinetic profiles and ATP production rates conducted with the Agilent Seahorse XFp Real-time ATP Rate assay are shown for (<b>C</b>) NPCs and (<b>D</b>) astrocytes. Dot plots display normalised mean ATP production rate ± SEM for NPCs and astrocytes using independent samples <span class="html-italic">t</span>-test or Mann–Whitney U test. Asterisks in the figure represent <span class="html-italic">p</span>-values as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Oxidative stress indicators in TSPO knockout and control cells. Cytosolic reactive oxygen species (ROS) and mitochondrial ROS (superoxide) were measured in (<b>A</b>) NPCs and (<b>B</b>) astrocytes by flow cytometry and are indicated by DCFDA and MitoSOX mean fluorescence intensity (MFI). Dot blots show MFI ± SEM using independent samples <span class="html-italic">t</span>-test of <span class="html-italic">n</span> = 4 individual experiments; 1 × 10<sup>5</sup> and 2 × 10<sup>4</sup> events were recorded for each replicate of NPCs and astrocytes, respectively. Histograms indicate the frequencies of measured values in the fluorescence intensity bins. Asterisks in the figure represent <span class="html-italic">p</span>-values as follows: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>(<b>A</b>) mtDNA copy number, (<b>B</b>) mitochondrial content, and (<b>C</b>) cell size in TSPO knockout and control cells. (<b>A</b>) mtDNA copy numbers in TSPO KO and CTRL cells are presented as single values of a minimum of three individual qRT-PCR of <span class="html-italic">n</span> = 9 (NPCs), <span class="html-italic">n</span> = 6 (astrocytes and neurons) biological replicates. (<b>B</b>) Mitochondrial content was measured by flow cytometry and is indicated by MitoTracker Green mean fluorescence intensity (MFI) of <span class="html-italic">n</span> = 4 individual experiments; 1 × 10<sup>5</sup> and 2 × 10<sup>4</sup> events were recorded for each replicate of NPCs and astrocytes, respectively. (<b>C</b>) Cell size was analysed by assessing the area [pixels] of Fura-2/AM-loaded cells, and the dot plots show the number of pixels. Data are presented as mean ± SEM using independent samples <span class="html-italic">t</span>-test or Mann–Whitney U test. (<b>D</b>–<b>G</b>) Relative gene and protein expression in TSPO knockout and control cells. (<b>D</b>) Gene expression of mitochondrial fusion and fission proteins <span class="html-italic">MFN1</span>, <span class="html-italic">OPA1</span>, and <span class="html-italic">DRP1</span>; (<b>E</b>) mitochondrial transcription factor <span class="html-italic">TFAM</span>; and (<b>F</b>) mitophagy proteins <span class="html-italic">PINK1</span> and <span class="html-italic">PARK2</span>. mRNA levels of TSPO-deficient cells related to TSPO-expressing control cells. <span class="html-italic">MFN1</span>: 0.9097, <span class="html-italic">p</span> = 0.0348; <span class="html-italic">OPA1</span>: 0.9393, <span class="html-italic">p</span> = 0.1989; <span class="html-italic">DRP1</span>: 0.9703, <span class="html-italic">p</span> = 0.6444; <span class="html-italic">TFAM</span>: 0.8581, <span class="html-italic">p</span> = 0.05; <span class="html-italic">PINK1</span>: 0.8401, <span class="html-italic">p</span> = 0.0988; <span class="html-italic">Parkin</span>: 0.6546, <span class="html-italic">p</span> = 0.0041. Data are presented as mean single values of five qRT-PCR ± SEM (<span class="html-italic">n</span> = 10), Welch’s corrected <span class="html-italic">t</span>-test. (<b>G</b>) Deletion of TSPO led to increased expression of the lipidated form of LC3B protein (LC3B-II) in hiPSC-derived NPCs and astrocytes. Representative Western blots show the expression of housekeeper gene GAPDH at 37 kDa and LC3B-I at 16 kDa and LC3B-II at 14 kDa. Bar graphs show densitometric, relative mean of LC3B-II expression levels. Individual values were normalised to the respective expression levels of GAPDH and were related to the CTRL mean. Data are presented as mean ± SEM using Mann–Whitney U test (Astro) and independent samples <span class="html-italic">t</span>-test (NPCs) of three biological replicates. (<b>H</b>,<b>I</b>) Analysis of the mitochondrial structure by electron microscopy in TSPO knockout and control astrocytes. (<b>H</b>) Representative images of mitochondria from astrocytes of both control and knockout groups (<span class="html-italic">n</span> = 3 each). (<b>I</b>) Semi-quantitative analysis of mitochondrial area, perimeter, and sphericity. Dot plots display single values of TSPO-expressing (<span class="html-italic">n</span> = 686) and TSPO-devoid (<span class="html-italic">n</span> = 854) mitochondria. Data are presented as mean ± SEM using Mann–Whitney U test. Asterisks in the figure represent <span class="html-italic">p</span>-values as follows: * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 8
<p>TSPO and VDAC1 protein expression in human primary fibroblasts of major depressive disorder (MDD) patients and healthy controls. Relative expression of (<b>A</b>) TSPO and (<b>B</b>) VDAC1 proteins normalised to the respective expression levels of BTUB1 and related to the CTRL mean. Bar graphs show densitometric, relative mean ± SEM using Mann–Whitney U test of three individual biological replicates of <span class="html-italic">n</span> = 16 fibroblast cell lines. Asterisks in the figure represent <span class="html-italic">p</span>-values as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
31 pages, 773 KiB  
Review
Skin and Induced Pluripotent Stem Cells as Biomarkers for Neurodegenerative Diseases
by Efstathios Rallis, Vasiliki-Sofia Grech, Kleomenis Lotsaris, Niki Tertipi, Eleni Sfyri and Vassiliki Kefala
Genes 2024, 15(12), 1507; https://doi.org/10.3390/genes15121507 - 25 Nov 2024
Viewed by 859
Abstract
As the global population ages, the rising prevalence of neurodegenerative diseases, characterized by abnormal protein aggregates, presents significant challenges for early diagnosis and disease monitoring. Identifying accessible tissue biomarkers is crucial for advancing our ability to detect and track the progression of these [...] Read more.
As the global population ages, the rising prevalence of neurodegenerative diseases, characterized by abnormal protein aggregates, presents significant challenges for early diagnosis and disease monitoring. Identifying accessible tissue biomarkers is crucial for advancing our ability to detect and track the progression of these diseases. Among the most promising biomarkers is the skin, which shares a common embryological origin with the brain and central nervous system (CNS). This biological connection positions the skin as a potential reflection of CNS pathology. Over the past decades, gene expression studies have demonstrated that key genes involved in neurodegenerative diseases are also expressed in skin tissues. Genes such as APP, PSEN1, PPA2, PINK1, LRRK2, PLCB4, MAPT, SPAST, and SPG7 are prominent in this regard. Beyond gene expression, proteins related to neurodegenerative diseases—such as α-synuclein, TAU, PARKIN, and prion protein (PrP)—have been isolated from the skin of affected individuals, underscoring the skin’s capacity to mirror neural degeneration. This non-invasive window into neurodegenerative processes is further enhanced by advances in stem cell technology, which have allowed for the generation of human-induced pluripotent stem cells (iPSCs) from patient-derived fibroblasts. These iPSCs offer a valuable model for studying disease mechanisms and developing therapeutic approaches. This review conducts a comprehensive analysis of the literature from databases such as PubMed, Google Scholar, and ResearchGate, emphasizing the unique potential of the skin as a non-invasive biomarker for neurodegenerative diseases. It explores how the skin serves as a bridge between gene expression and disease pathology in both the skin and the CNS. By leveraging this biological connection, the skin emerges as a promising model for enhancing our understanding of neurodegenerative disorders and developing innovative strategies for early detection and treatment. However, significant limitations remain, requiring further validation to establish the specificity and sensitivity of these biomarkers. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Figure 1
<p>Integration of skin fibroblasts and iPSC technology in neurodegenerative disease research.</p>
Full article ">
15 pages, 7842 KiB  
Article
Human-Induced Pluripotent Stem Cell-Derived Neural Organoids as a Novel In Vitro Platform for Developmental Neurotoxicity Assessment
by Tsunehiko Hongen, Kenta Sakai, Tomohiro Ito, Xian-Yang Qin and Hideko Sone
Int. J. Mol. Sci. 2024, 25(23), 12523; https://doi.org/10.3390/ijms252312523 - 21 Nov 2024
Viewed by 1254
Abstract
There has been a recent drive to replace in vivo studies with in vitro studies in the field of toxicity testing. Therefore, instead of conventional animal or planar cell culture models, there is an urgent need for in vitro systems whose conditions can [...] Read more.
There has been a recent drive to replace in vivo studies with in vitro studies in the field of toxicity testing. Therefore, instead of conventional animal or planar cell culture models, there is an urgent need for in vitro systems whose conditions can be strictly controlled, including cell–cell interactions and sensitivity to low doses of chemicals. Neural organoids generated from human-induced pluripotent stem cells (iPSCs) are a promising in vitro platform for modeling human brain development. In this study, we developed a new tool based on various iPSCs to study and predict chemical-induced toxicity in humans. The model displayed several neurodevelopmental features and showed good reproducibility, comparable to that of previously published models. The results revealed that basic fibroblast growth factor plays a key role in the formation of the embryoid body, as well as complex neural networks and higher-order structures such as layered stacking. Using organoid models, pesticide toxicities were assessed. Cells treated with low concentrations of rotenone underwent apoptosis to a greater extent than those treated with high concentrations of rotenone. Morphological changes associated with the development of neural progenitor cells were observed after exposure to low doses of chlorpyrifos. These findings suggest that the neuronal organoids developed in this study mimic the developmental processes occurring in the brain and nerves and are a useful tool for evaluating drug efficacy, safety, and toxicity. Full article
(This article belongs to the Special Issue Environmental Epigenome and Endocrine Disrupting Chemicals)
Show Figures

Figure 1

Figure 1
<p>Immunostaining images depicting the effect of bFGF on organoid differentiation at various stages. (<b>A</b>–<b>C</b>) present immunostained images of organoids on days 24, 34, and 50, respectively. (<b>D</b>) presents a magnified view of the upper part of the bFGF-exposed organoid on day 50. Staining included Hoechst staining (in blue) for nuclear labeling, neuron markers (in green) for neuron identification, and phalloidin–rhodamine (in red) for cytoskeletal visualization. Images were captured using Olympus FV10-ASW (10× objective; Scale bar, 200 μm).</p>
Full article ">Figure 2
<p>Forebrain organoid differentiation protocol. Cells were seeded at a density of 9000 cells per well, and embryoid bodies were allowed to form until day 4 of culture. The cells were then naturally induced in SMAD (Sma and Mad related protein) inhibitor-containing medium until day 15, differentiated into forebrain-type organoids from day 16, and cultured to maturity from days 22 to 29. Maintenance culture was continued until day 50.</p>
Full article ">Figure 3
<p>Fluorescence immunostaining of forebrain-type organoids on day 50 of culture. (<b>A</b>) Internal structure of the organoids on day 50 of culture, as observed using a confocal microscope Z-stack with a 25× objective. (<b>B</b>) Magnified image using a 40× objective. (<b>C</b>–<b>E</b>) Localization of cells by each marker. Cell nuclei (blue), neurons (green), and cytoskeleton (red) are shown (4× objective; Scale bar, 100 μm. (<b>F</b>) A forebrain organoid stained with the glial cell marker GFAP (red) and neurons (green). (<b>F</b>,<b>G</b>) An enlarged view of the upper part (4× objective; Scale bar, 200 μm).</p>
Full article ">Figure 4
<p>RNA sequencing analysis of human-induced pluripotent stem cells exposed to rotenone. (<b>A</b>) Heatmap illustrating the differential gene expression between control and rotenone-treated samples. Genes are ranked based on their standard deviation across all samples, and the top 1000 genes are used for hierarchical clustering analysis. Details of which genes are upregulated or downregulated in the control versus rotenone treatment are provided in the table within the <a href="#app1-ijms-25-12523" class="html-app">Supplemental Materials</a>. (<b>B</b>) The data shows network interactions in the KEGG signaling pathways. In particular, focal adhesion and the neuroactive ligand–receptor interactions interact largely the others. (<b>C</b>) Analysis of gene pathways with high variability for up- and down-regulation of gene expressions.</p>
Full article ">Figure 4 Cont.
<p>RNA sequencing analysis of human-induced pluripotent stem cells exposed to rotenone. (<b>A</b>) Heatmap illustrating the differential gene expression between control and rotenone-treated samples. Genes are ranked based on their standard deviation across all samples, and the top 1000 genes are used for hierarchical clustering analysis. Details of which genes are upregulated or downregulated in the control versus rotenone treatment are provided in the table within the <a href="#app1-ijms-25-12523" class="html-app">Supplemental Materials</a>. (<b>B</b>) The data shows network interactions in the KEGG signaling pathways. In particular, focal adhesion and the neuroactive ligand–receptor interactions interact largely the others. (<b>C</b>) Analysis of gene pathways with high variability for up- and down-regulation of gene expressions.</p>
Full article ">Figure 5
<p>Typical morphological changes in forebrain organoids induced by the pesticides rotenone and chlorpyrifos. (<b>A</b>,<b>B</b>) Rotenone. (<b>C</b>,<b>D</b>) Chlorpyrifos. Morphological changes and respective size changes from day 1 to day 11 of incubation. ((<b>A</b>,<b>C</b>) 4× objective; Scale bar, 2.0 mm). Experiments were performed independently in triplicate or quadruplicate. Asterisks indicate significant differences between the exposed and control groups <span class="html-italic">p</span> &lt; 0.05 = *; <span class="html-italic">p</span> &lt; 0.01 = **. N = 4.</p>
Full article ">Figure 6
<p>Typical morphological changes in 29-day pre-brain organoids in culture induced by the pesticides rotenone and chlorpyrifos. (<b>A</b>,<b>B</b>) Rotenone. (<b>C</b>,<b>D</b>) Chlorpyrifos. Morphological changes and respective size changes on day 29 of incubation. ((<b>A</b>,<b>C</b>); 4× objective; Scale bar, 2.0 mm). Experiments were performed independently in triplicate or quadruplicate. N = 4.</p>
Full article ">Figure 7
<p>Immunofluorescence images of forebrain-type organoids at 29 days in culture to visualize the effects of chemical substances on neural cell development. (<b>A</b>) Internal structure of forebrain-type organoids in the control group, captured in two fields of view (10× and 20× objective). The red boxes indicate nerve dense foci, while the white boxes indicate winding structures. (<b>B</b>) Internal structure of organoids exposed to 10 μM rotenone. (<b>C</b>) Internal structure of organoids exposed to 10 μM chlorpyrifos. Cell nuclei (blue), neurons (green), and glial cells (red) are shown (<span class="html-italic">n</span> = 3; 10× objective; scale bar, 200 μm and 20× objective; scale bar, 100 μm).</p>
Full article ">Figure 8
<p>The morphological characteristics of the forebrain organoids identified in <a href="#ijms-25-12523-f007" class="html-fig">Figure 7</a> were evaluated. (<b>A</b>) 1. Neurites were scored as 1 point; 2. nerve dense foci as 2 points; and 3. The red dotted lines indicate three points of the winding structures (scale bar, 2.0 mm). (<b>B</b>) Morphological evaluation scores (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 9
<p>Gene expression analysis of forebrain organoids exposed to rotenone and chlorpyrifos. (<b>A</b>) Changes in the expression levels of neuronal marker genes (MAP2, SOX2, PAX6) and apoptosis-related marker genes (ADM, CASP3, CASP7) following exposure to various concentrations of rotenone. (<b>B</b>) Changes in the expression levels of neuronal and apoptosis marker genes following exposure to chlorpyrifos. Results are presented as the mean ± SD (<span class="html-italic">n</span> = 3). Asterisks indicate <span class="html-italic">p</span>-values based on multiple comparisons via two-way analysis of variance <span class="html-italic">p</span> &lt; 0.05 = *; <span class="html-italic">p</span> &lt; 0.01 = **. The red dotted lines indicate the control value.</p>
Full article ">
31 pages, 12498 KiB  
Review
Using Small Molecules to Reprogram RPE Cells in Regenerative Medicine for Degenerative Eye Disease
by Lyubov A. Rzhanova, Elena V. Alpeeva and Maria A. Aleksandrova
Cells 2024, 13(23), 1931; https://doi.org/10.3390/cells13231931 - 21 Nov 2024
Viewed by 587
Abstract
The main purpose of regenerative medicine for degenerative eye diseases is to create cells to replace lost or damaged ones. Due to their anatomical, genetic, and epigenetic features, characteristics of origin, evolutionary inheritance, capacity for dedifferentiation, proliferation, and plasticity, mammalian and human RPE [...] Read more.
The main purpose of regenerative medicine for degenerative eye diseases is to create cells to replace lost or damaged ones. Due to their anatomical, genetic, and epigenetic features, characteristics of origin, evolutionary inheritance, capacity for dedifferentiation, proliferation, and plasticity, mammalian and human RPE cells are of great interest as endogenous sources of new photoreceptors and other neurons for the degrading retina. Promising methods for the reprogramming of RPE cells into retinal cells include genetic methods and chemical methods under the influence of certain low-molecular-weight compounds, so-called small molecules. Depending on the goal, which can be the preservation or the replacement of lost RPE cells and cellular structures, various small molecules are used to influence certain biological processes at different levels of cellular regulation. This review discusses the potential of the chemical reprogramming of RPE cells in comparison with other somatic cells and induced pluripotent stem cells (iPSCs) into neural cells of the brain and retina. Possible mechanisms of the chemically induced reprogramming of somatic cells under the influence of small molecules are explored and compared. This review also considers other possibilities in using them in the treatment of retinal degenerative diseases based on the protection, preservation, and support of survived RPE and retinal cells. Full article
(This article belongs to the Special Issue Mechanism of Cell Signaling during Eye Development and Diseases)
Show Figures

Figure 1

Figure 1
<p>Eye injury induces different RPE responses in lower vertebrates and mammals, including humans. The initial stages of cellular reprogramming are the same in all studied organisms: cells enter the cell cycle, begin to proliferate, lose pigment, and dedifferentiate. However, subsequently, the cells develop completely differently: in some species, RPE cells are transformed into neuronal retinal cells, thereby restoring the retina, while in humans, dedifferentiated RPE cells differentiate into myofibroblasts, which leads to serious pathologies.</p>
Full article ">Figure 2
<p>Some small molecules and signaling pathways promoting the development of the nervous system (based on the poster <a href="https://www.stemcell.com/media/files/wallchart/WA10014-Small_Molecules_Big_Impact.pdf" target="_blank">https://www.stemcell.com/media/files/wallchart/WA10014-Small_Molecules_Big_Impact.pdf</a> (accessed on 16 November 2024)). Notes: The transforming growth factor β (TGFβ) is involved in a whole range of biological functions, from cell growth to cell differentiation and apoptosis. SMAD1, 2, 3, 5, and 8 are receptor-regulated SMADs. They bind to membrane-bound serine/threonine receptors and are activated by the kinase activity of the receptors. SMAD4 acts as a cofactor that binds to activated R-SMADS (SMADs) forming a complex that translocates into the nucleus [<a href="#B63-cells-13-01931" class="html-bibr">63</a>]. Pathway inhibitors: SB431542, LY364947, RepSox, Dorsomorphin, LDN193189. The Notch signaling pathway regulates cell proliferation, cell fate, differentiation, and cell death in all metazoans. The Notch pathway is activated when Delta or Jagged ligands on neighboring cells activate cleavage of the receptor releasing the Notch intracellular domain (NICD). The Notch pathway plays a role in specifying neural subtypes [<a href="#B64-cells-13-01931" class="html-bibr">64</a>]. Pathway inhibitors: DAPT, LY411575. Fibroblast growth factor (FGF) signaling regulates several developmental processes, including cellular proliferation, differentiation, migration, morphogenesis, and patterning. FGF signaling via MEK/ERK is critical for self-renewal and proliferation of human PSCs [<a href="#B65-cells-13-01931" class="html-bibr">65</a>]. The WNT signaling pathway is an ancient and evolutionarily conserved pathway that regulates crucial aspects of cell fate determination, cell migration, cell polarity, neural patterning, and organogenesis during embryonic development [<a href="#B66-cells-13-01931" class="html-bibr">66</a>]. Pathway activators: CHIR99021, SB216763; pathway inhibitors: IWR-1-endo. The Hedgehog (Shh) pathway is important in post-embryonic tissue regeneration and repair processes. Specifically, Shh signaling is implicated in the induction of multifarious neuronal populations in central nervous system [<a href="#B67-cells-13-01931" class="html-bibr">67</a>]. Pathway activators: Purmorphamine, SAg. The RHO/ROCK pathway regulates cytoskeletal dynamics and plays an important role in cell adhesion, proliferation, motility, contraction, and apoptosis. Loss of cadherin or integrin binding activates the Rho pathway in human PSCs, leading to anoikis [<a href="#B68-cells-13-01931" class="html-bibr">68</a>]. Pathway inhibitors: Y-27632, thiazovivin. The 3′,5′-cyclic adenosine monophosphate (cAMP) is a second messenger important in reprogramming and differentiation for many cell subtypes [<a href="#B69-cells-13-01931" class="html-bibr">69</a>]. Pathway activator: forskolin. The protein kinase C (PKC) family of kinases is commonly activated by diacylglycerol (DAG) and calcium and is involved in several signaling pathways that can regulate differentiation [<a href="#B70-cells-13-01931" class="html-bibr">70</a>]. Pathway activators: prostaglandin E2, (−)-Indolactam V; pathway inhibitors: HA-100, GO6983. Retinoic acid (RA) is a potent morphogen required for embryonic development. RA acts in a paracrine fashion to shape the developing eye and is essential for normal optic vesicle and anterior segment formation [<a href="#B71-cells-13-01931" class="html-bibr">71</a>]. Activators: 9-cis retinoic acid, all-trans retinoic acid, CD437, TTNPB. RAR, RXR -RA receptors. Epigenetic marks such as acetylation (Ac) of histones and methylation (Me) of histones or DNA serve to induce or inhibit gene expression in a heritable manner. Global changes in epigenetic marks are critical for reprogramming [<a href="#B35-cells-13-01931" class="html-bibr">35</a>]. DNA Methyltransferase inhibitors: RG108; histone methyltransferase inhibitors: BIX01294; histone demethylase inhibitors: tranylcypromine; histone acetyltransferase inhibitors: garcinol; histone deacetylase inhibitors: sodium butyrate, trichostatin A, valproic acid.</p>
Full article ">Figure 3
<p>Schematic representation of chemically induced reprogramming of fibroblasts into neural stem cells and into neurons in the brain and retina [<a href="#B12-cells-13-01931" class="html-bibr">12</a>,<a href="#B36-cells-13-01931" class="html-bibr">36</a>,<a href="#B37-cells-13-01931" class="html-bibr">37</a>,<a href="#B39-cells-13-01931" class="html-bibr">39</a>,<a href="#B41-cells-13-01931" class="html-bibr">41</a>,<a href="#B42-cells-13-01931" class="html-bibr">42</a>,<a href="#B43-cells-13-01931" class="html-bibr">43</a>,<a href="#B85-cells-13-01931" class="html-bibr">85</a>,<a href="#B86-cells-13-01931" class="html-bibr">86</a>]. This approach utilized small molecules that acted on the cellular epigenome (Epi) and on various signaling pathways that control cellular identity (TGFβ, GSK3β, PKC, BMP, SHH, JNR, ROCK, and others). FG—growth factor; m—mouse; h—human; CiNs—chemically induced neurons; CiNSCs—chemically induced neural stem cells; CiPCs—chemically induced photoreceptor-like cells; m rd1—mouse model of retinal degeneration; m NaIO<sub>3</sub>—mouse model of sodium iodate (NaIO<sub>3</sub>)-induced retinal degeneration; SRT—subretinal transplantation, LVT—lateral ventricle transplantation. Pathway activators: green color; pathway inhibitors: red color.</p>
Full article ">Figure 4
<p>Schematic representation of chemically induced reprogramming of astrocytes into neural stem cells and into neurons in the brain and retina [<a href="#B34-cells-13-01931" class="html-bibr">34</a>,<a href="#B38-cells-13-01931" class="html-bibr">38</a>,<a href="#B50-cells-13-01931" class="html-bibr">50</a>,<a href="#B87-cells-13-01931" class="html-bibr">87</a>,<a href="#B88-cells-13-01931" class="html-bibr">88</a>,<a href="#B89-cells-13-01931" class="html-bibr">89</a>,<a href="#B90-cells-13-01931" class="html-bibr">90</a>,<a href="#B91-cells-13-01931" class="html-bibr">91</a>,<a href="#B92-cells-13-01931" class="html-bibr">92</a>]. Chemically induced reprogramming utilized small molecules that acted on the cellular epigenome (Epi) and on various signaling pathways that control cellular identity (TGFβ, GSK3β, PKC, SHH, Notch, RAR, ROCK, and others). FG—growth factor; m—mouse; nm—neonatal mouse; h—human; r—rat; CiNs—chemically induced neurons; CiNSCs—chemically induced neural stem cells; CRI—microinjection into the cortices; STI—microinjection into the striatum; LVT—lateral ventricle transplantation. Pathway activators: green color; pathway inhibitors: red color.</p>
Full article ">Figure 5
<p>Schematic representation of chemically induced reprogramming of ESCs and iPSCs into neural stem cells and into neurons of the brain and retina [<a href="#B2-cells-13-01931" class="html-bibr">2</a>,<a href="#B47-cells-13-01931" class="html-bibr">47</a>,<a href="#B52-cells-13-01931" class="html-bibr">52</a>,<a href="#B54-cells-13-01931" class="html-bibr">54</a>,<a href="#B97-cells-13-01931" class="html-bibr">97</a>,<a href="#B98-cells-13-01931" class="html-bibr">98</a>,<a href="#B99-cells-13-01931" class="html-bibr">99</a>]. Chemically induced reprogramming utilized small molecules that acted on the cellular epigenome (Epi) and on various signaling pathways that control cellular identity (TGFβ, GSK3β, WNT, BMP, NOTCH, ROCK, and others). GF—growth factor; iPSC—induced pluripotent stem cell; ESCs—embryonic stem cells; RSCs—retinal stem cell; CiRGCs—chemically induced retinal ganglion cells; CiR—chemically induced retina; CiROD—chemically induced rods; CiRPE—chemically induced retinal pigment epithelium; CiPCs—chemically induced photoreceptor-like cells; m NOD-SCID—the nonobese diabetic/severe combined immunodeficient mouse; m Crx<sup>tvrm65</sup>/IL2rγ<sup>−/−</sup>—model of immunosuppressive mouse/retinal degeneration; m NOD.SCID-rd1—the nonobese diabetic/severe combined immunodeficient mouse model of retinal degeneration; RCS rat—rat model of retinal degeneration from Royal College of Surgeons; IVT—intravitreal injection; SRT—subretinal injection; mMNU—mouse model of N-Nitroso-N-methylurea (MNU)-induced retinal degeneration, m—mouse, h—human. Pathway activators: green color; pathway inhibitors: red color. 2.3.4. Reprogramming of the RPE into CiNSCs and CiNs.</p>
Full article ">Figure 6
<p>Schematic representation of chemically induced reprogramming of RPE into neural stem cells and into neurons in the brain and retina [<a href="#B12-cells-13-01931" class="html-bibr">12</a>,<a href="#B27-cells-13-01931" class="html-bibr">27</a>,<a href="#B46-cells-13-01931" class="html-bibr">46</a>,<a href="#B93-cells-13-01931" class="html-bibr">93</a>]. Chemically induced reprogramming utilized small molecules that acted on the cellular epigenome (Epi) and on various signaling pathways that control cellular identity (TGFβ, GSK3β, WNT, BMP, NOTCH, PKC, and others). Fh—fetal human; Mn—cynomolgus monkeys (<span class="html-italic">Macaca fascicularis</span>); MPTP hydrochloride—induced Parkinson’s disease model; PPI—implantation into posterior putamen; sphere—free floating conditions; for other abbreviations, refer to <a href="#cells-13-01931-f003" class="html-fig">Figure 3</a> and <a href="#cells-13-01931-f005" class="html-fig">Figure 5</a>. Pathway activators: green color; pathway inhibitors: red color.</p>
Full article ">
12 pages, 5784 KiB  
Article
Tlx Promotes Stroke-Induced Neurogenesis and Neuronal Repair in Young and Aged Mice
by Dilaware Khan, Dagmar Bock, Hai-Kun Liu and Sajjad Muhammad
Int. J. Mol. Sci. 2024, 25(22), 12440; https://doi.org/10.3390/ijms252212440 - 19 Nov 2024
Viewed by 634
Abstract
Stroke is one of the leading causes of chronic disability in humans. It has been proposed that the endogenous neural stem/progenitor cells generate new neurons in the damaged area. Still, the contribution of these cells is negligible because a low number of newborn [...] Read more.
Stroke is one of the leading causes of chronic disability in humans. It has been proposed that the endogenous neural stem/progenitor cells generate new neurons in the damaged area. Still, the contribution of these cells is negligible because a low number of newborn mature neurons are formed. Tlx conventional knock-out mice, Tlx-CreERT2 mice, and Tlx-overexpressing (Tlx-OE) mice were specifically chosen for their unique genetic characteristics, which were crucial for the experiments. Permanent and transient middle cerebral artery occlusion was used to induce stroke in the mice. Immunostainings for doublecortin and GFP/BrdU/NeuN were performed to study neurogenesis and fate mapping. The rotarod test was performed to assess motor deficits. Here, we show that stroke-induced neurogenesis is dramatically increased with the additional expression of two copies of the nuclear receptor-coding gene tailless (Tlx, also known as Nr2e1), which has been shown to be a master regulator of subventricular zone (SVZ) neural stem cells (NSCs). We show that Tlx expression is upregulated after stroke, and stroke-induced neurogenesis is blocked when Tlx is inactivated. Tlx overexpression in NSCs leads to massive induction of neurogenesis via stroke. More newborn mature neurons are formed in Tlx-overexpressing mice, leading to improved coordination and motor function recovery. Most importantly, we also demonstrate that this process is sustained in aged mice, where stroke-induced neurogenesis is nearly undetectable in wild-type animals. This study provides the first stem cell-specific genetic evidence that endogenous NSCs can be exploited by manipulating their master regulator, Tlx, and thus suggests a novel therapeutic strategy for neuronal repair. Full article
(This article belongs to the Special Issue Advances in Research on Neurogenesis: 3rd Edition)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Tlx</span> and <span class="html-italic">Tlx</span>-expressing cells are responsible for stroke-induced neurogenesis. (<b>a</b>) <span class="html-italic">Tlx</span> antibody staining of brain sections from sham (left) or stroke (right) WT mice 1 week after operation. (<b>b</b>) DCX antibody staining of brain sections from <span class="html-italic">Tlx<sup>−/−</sup></span> and control (<span class="html-italic">Tlx</span><sup>+/−</sup>) mice 1 week after stroke; arrows indicate individual DCX-positive cells in the striatum. Note that no DCX-positive cells are seen in the <span class="html-italic">Tlx<sup>−/−</sup></span> brain sections. (<b>c</b>) GFP antibody staining of brain sections from <span class="html-italic">Tlx</span>-CreER<sup>T2</sup>;Z/EG mice treated as shown by the scheme. (<b>d</b>) GFP/NeuN/BrdU triple staining of brain sections from <span class="html-italic">Tlx</span>-CreER<sup>T2</sup>;Z/EG mice treated as shown by the scheme. Scale bar: 20 μm.</p>
Full article ">Figure 2
<p><span class="html-italic">Tlx</span> overexpression expands stroke-induced neurogenesis. (<b>a</b>) DCX antibody staining of brain sections from <span class="html-italic">Tlx</span>-OE mice 2 weeks after stroke. Note that only a few DCX-positive cells are found in WT brain sections. (<b>b</b>) Number of DCX-positive cells per striatum of brain sections from the stroke side (<span class="html-italic">n</span> = 6, mean ± standard deviation (SD), ** <span class="html-italic">p</span> &lt; 0.01). (<b>c</b>) An overview of DCX staining in the <span class="html-italic">Tlx</span>-OE mouse brain sections. Note that stroke-induced neurogenesis is restricted to the stroke side (arrow) but not the collateral side. (<b>d</b>) BrdU IHC of brain sections from mice with stroke. BrdU was administrated daily for 5 days immediately after stroke. IHC was performed 2 days after the last BrdU injection. Note that more BrdU-positive cells are found in the striatum of <span class="html-italic">Tlx</span>-OE brain sections. (<b>e</b>) DCX staining of brain sections from <span class="html-italic">Tlx</span>-OE mice 2 weeks after stroke, arrows indicate clusters of DCX-positive cells. Scale bar: 20 μm.</p>
Full article ">Figure 3
<p><span class="html-italic">Tlx</span> overexpression-mediated increase in stroke-induced neurogenesis results in increase in newly formed mature neurons. (<b>A</b>) Scheme of experimental procedure. (<b>B</b>) BrdU/NeuN costaining of mouse brain sections 2 months after stroke. Mice received continuous BrdU injection for 5 days 1 week after stroke. Scale bar: 50 µm. (<b>C</b>) Number of BrdU/NeuN double-positive cells per section in the cortex 2 months after stroke (<span class="html-italic">n</span> = 8, mean ± SD, ** <span class="html-italic">p</span> &lt; 0.01). (<b>D</b>) Z-stack image of two adjacent BrdU/NeuN double-positive cells in the <span class="html-italic">Tlx</span>-OE cortex 2 months after stroke. Scale bar: 20 µm. (<b>E</b>) Rotarod test demonstrates that <span class="html-italic">Tlx</span>-OE mice recover better than control animals after stroke. (<span class="html-italic">n</span> = 14, * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p><span class="html-italic">Tlx</span> overexpression results in consistent stroke-induced neurogenic phenotype in aged mice. (<b>a</b>) DCX IHC of stroke brain sections (2 weeks) from 1 year old mice. Note that DCX-positive cells in the WT striatum are very rare. Scale bar: 50 µm. (<b>b</b>) DCX staining on sagittal sections of 1 year old mice 2 months after stroke; pictures acquired from the square region indicate the initial part of the rostral migratory stream. (<b>c</b>) Overview of DCX staining pattern described above. Note that the majority of DCX-positive cells are orientated toward the lesion area. (Inset showing a higher magnification). Scale bar: 100 µm. (<b>d</b>) BrdU/NeuN costaining of brain sections of approximately 1-year-old mice 3 months after stroke. Scale bar: 20 µm. (<b>e</b>) Number of BrdU/NeuN-double positive cells in striatum 3 months after stroke (<span class="html-italic">n</span> = 6, mean ± SD, * <span class="html-italic">p</span> &lt; 0.05). (<b>f</b>) Z-stack image of a BrdU/NeuN double-positive cell in the <span class="html-italic">Tlx</span>-OE cortex 3 months after stroke. Scale bar: 20 µm.</p>
Full article ">
19 pages, 3396 KiB  
Article
A Snapshot of Early Transcriptional Changes Accompanying the Pro-Neural Phenotype Switch by NGN2, ASCL1, SOX2, and MSI1 in Human Fibroblasts: An RNA-Seq Study
by Ekaterina M. Samoilova, Daria A. Chudakova, Erdem B. Dashinimaev, Anastasiya V. Snezhkina, Olga M. Kudryashova, Anastasia V. Lipatova, Alesya V. Soboleva, Pavel O. Vorob’yev, Vladimir T. Valuev-Elliston, Natalia F. Zakirova, Alexander V. Ivanov and Vladimir P. Baklaushev
Int. J. Mol. Sci. 2024, 25(22), 12385; https://doi.org/10.3390/ijms252212385 - 18 Nov 2024
Viewed by 722
Abstract
Direct pro-neural reprogramming is a conversion of differentiated somatic cells to neural cells without an intermediate pluripotency stage. It is usually achieved via ectopic expression (EE) of certain transcription factors (TFs) or other reprogramming factors (RFs). Determining the transcriptional changes (TCs) caused by [...] Read more.
Direct pro-neural reprogramming is a conversion of differentiated somatic cells to neural cells without an intermediate pluripotency stage. It is usually achieved via ectopic expression (EE) of certain transcription factors (TFs) or other reprogramming factors (RFs). Determining the transcriptional changes (TCs) caused by particular RFs in a given cell line enables an informed approach to reprogramming initiation. Here, we characterized TCs in the human fibroblast cell line LF1 on the 5th day after EE of the single well-known pro-neural RFs NGN2, ASCL1, SOX2, and MSI1. As assessed by expression analysis of the bona fide neuronal markers nestin and beta-III tubulin, all four RFs initiated pro-neuronal phenotype conversion; analysis by RNA-seq revealed striking differences in the resulting TCs, although some pathways were overlapping. ASCL1 and SOX2 were not sufficient to induce significant pro-neural phenotype switches using our EE system. NGN2 induced TCs indicative of cell phenotype changes towards neural crest cells, neural stem cells, mature neurons, as well as radial glia, astrocytes, and oligodendrocyte precursors and their mature forms. MSI1 mainly induced a switch towards early stem-like cells, such as radial glia. Full article
(This article belongs to the Special Issue Current Molecular Progress on Cell and Gene Therapies)
Show Figures

Figure 1

Figure 1
<p>Immunocytochemical analysis of LF1 cells on day 5 after transduction. Confocal microscopy images of cells stained for nestin, β-III-tubulin, and GFAP. LF1-ASCL1 (<b>A</b>–<b>C</b>), LF1-SOX2 (<b>D</b>–<b>F</b>), LF1-MSI1 (<b>G</b>–<b>I</b>), LF1-NGN2 (<b>J</b>–<b>L</b>) samples. Green channel: GFP fluorescence confirming the integration of the lentiviral vectors. Red channel: fluorescence of the secondary antibodies (Alexa Fluor 633). The cell nuclei are stained with Hoechst (blue) in all the panels.</p>
Full article ">Figure 2
<p>Differential expression analysis of RNA-seq data (<b>A</b>) Principal component analysis (PCA) of the log2 RNA-seq data presented as a two-dimensional scatterplot of the first two principal components. The sample groups are represented by different colors, each dot represents a biological replicate; the sample legend is provided within the plot; (<b>B</b>,<b>C</b>) Venn diagrams of the DEGs across LF1-NGN2, LF1-MSI1, and LF1-LeGoiG2 that were up- (<b>B</b>) and down- (<b>C</b>) regulated. (<b>D</b>–<b>G</b>) For LF1-NGN2 and LF1-MSI1, based on each TF’s targets amongst the DEGs, the top 25 most highly affected TFs are shown ((<b>D</b>,<b>F</b>), correspondingly), as well as the estimated pathway activity ((<b>E</b>,<b>G</b>), correspondingly).</p>
Full article ">Figure 3
<p>Profiles of cell subtypes in the samples based on gene expression analysis. Heatmap showing enrichment of the samples with DEGs of pro-neuronal cell populations (<b>A</b>), stem-like cells (<b>B</b>), neural crest cells (<b>C</b>), and radial glial cells (<b>D</b>). F, control fibroblasts LF1, FL, LF-LeGoiG2-Puro+, FLA, LF-LeGoiG2-Puro+-ASCL1, FLM, LF-LeGoiG2-Puro+-MSI1, FLN, LF-LeGoiG2-Puro+-NGN2, FLS, LF-LeGoiG2-Puro+-SOX2. Blue and red color scale represents the median-scaled change in gene expression or cell population enrichment (red—up, blue—down).</p>
Full article ">Figure 4
<p>Profiles of cell subtypes in the samples based on gene expression analysis. Heatmap showing enrichment of the samples with DEGs of neural progenitors (<b>A</b>), neurons (<b>B</b>), presynaptic (<b>C</b>), and postsynaptic structures (<b>D</b>). F, control fibroblasts LF1, FL, LF-LeGoiG2-Puro+, FLA, LF-LeGoiG2-Puro+-ASCL1, FLM, LF-LeGoiG2-Puro+-MSI1, FLN, LF-LeGoiG2-Puro+-NGN2, FLS, LF-LeGoiG2-Puro+-SOX2. Blue and red color scale represents the median-scaled change in gene expression or cell population enrichment (red—up, blue—down).</p>
Full article ">Figure 5
<p>Profiles of cell subtypes in the samples based on gene expression analysis. Heatmap showing enrichment of the samples with DEGs of astrocytes (<b>A</b>) and oligodendrocyte precursors (<b>B</b>). F, control fibroblasts LF1, FL, LF-LeGoiG2-Puro+, FLA, LF-LeGoiG2-Puro+-ASCL1, FLM, LF-LeGoiG2-Puro+-MSI1, FLN, LF-LeGoiG2-Puro+-NGN2, FLS, LF-LeGoiG2-Puro+-SOX2. Blue and red color scale represents the median-scaled change in gene expression or cell population enrichment (red—up, blue—down).</p>
Full article ">
17 pages, 711 KiB  
Review
Proposed Mechanisms of Cell Therapy for Alzheimer’s Disease
by Ekaterina Belousova, Diana Salikhova, Yaroslav Maksimov, Vladimir Nebogatikov, Anastasiya Sudina, Dmitry Goldshtein and Aleksey Ustyugov
Int. J. Mol. Sci. 2024, 25(22), 12378; https://doi.org/10.3390/ijms252212378 - 18 Nov 2024
Viewed by 853
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder characterized by mitochondria dysfunction, accumulation of beta-amyloid plaques, and hyperphosphorylated tau tangles in the brain leading to memory loss and cognitive deficits. There is currently no cure for this condition, but the potential of stem cells [...] Read more.
Alzheimer’s disease is a progressive neurodegenerative disorder characterized by mitochondria dysfunction, accumulation of beta-amyloid plaques, and hyperphosphorylated tau tangles in the brain leading to memory loss and cognitive deficits. There is currently no cure for this condition, but the potential of stem cells for the therapy of neurodegenerative pathologies is actively being researched. This review discusses preclinical and clinical studies that have used mouse models and human patients to investigate the use of novel types of stem cell treatment approaches. The findings provide valuable insights into the applications of stem cell-based therapies and include the use of neural, glial, mesenchymal, embryonic, and induced pluripotent stem cells. We cover current studies on stem cell replacement therapy where cells can functionally integrate into neural networks, replace damaged neurons, and strengthen impaired synaptic circuits in the brain. We address the paracrine action of stem cells acting via secreted factors to induce neuroregeneration and modify inflammatory responses. We focus on the neuroprotective functions of exosomes as well as their neurogenic and synaptogenic effects. We look into the shuttling of mitochondria through tunneling nanotubes that enables the transfer of healthy mitochondria by restoring the normal functioning of damaged cells, improving their metabolism, and reducing the level of apoptosis. Full article
(This article belongs to the Special Issue From Molecular Insights to Novel Therapies: Neurological Diseases)
Show Figures

Figure 1

Figure 1
<p>Stem cell strategies for Alzheimer’s disease and proposed mechanisms of stem cell therapy. Mesenchymal stem cells (MSCs), neural stem cells (NSCs), glial progenitor cells (GPCs), neural progenitor cells (NPCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs) participate in various types of cell processing affecting cell replacement and are involved in paracrine action via secreted factors and exosomes, as well as the transfer of mitochondria through tunneling nanotube (TNT) pathway.</p>
Full article ">
28 pages, 2534 KiB  
Review
NMDA Receptors in Neurodevelopmental Disorders: Pathophysiology and Disease Models
by Roshan Tumdam, Yara Hussein, Tali Garin-Shkolnik and Shani Stern
Int. J. Mol. Sci. 2024, 25(22), 12366; https://doi.org/10.3390/ijms252212366 - 18 Nov 2024
Viewed by 1621
Abstract
N-methyl-D-aspartate receptors (NMDARs) are critical components of the mammalian central nervous system, involved in synaptic transmission, plasticity, and neurodevelopment. This review focuses on the structural and functional characteristics of NMDARs, with a particular emphasis on the GRIN2 subunits (GluN2A-D). The diversity of GRIN2 [...] Read more.
N-methyl-D-aspartate receptors (NMDARs) are critical components of the mammalian central nervous system, involved in synaptic transmission, plasticity, and neurodevelopment. This review focuses on the structural and functional characteristics of NMDARs, with a particular emphasis on the GRIN2 subunits (GluN2A-D). The diversity of GRIN2 subunits, driven by alternative splicing and genetic variants, significantly impacts receptor function, synaptic localization, and disease manifestation. The temporal and spatial expression of these subunits is essential for typical neural development, with each subunit supporting distinct phases of synaptic formation and plasticity. Disruptions in their developmental regulation are linked to neurodevelopmental disorders, underscoring the importance of understanding these dynamics in NDD pathophysiology. We explore the physiological properties and developmental regulation of these subunits, highlighting their roles in the pathophysiology of various NDDs, including ASD, epilepsy, and schizophrenia. By reviewing current knowledge and experimental models, including mouse models and human-induced pluripotent stem cells (hiPSCs), this article aims to elucidate different approaches through which the intricacies of NMDAR dysfunction in NDDs are currently being explored. The comprehensive understanding of NMDAR subunit composition and their mutations provides a foundation for developing targeted therapeutic strategies to address these complex disorders. Full article
Show Figures

Figure 1

Figure 1
<p>Structural composition of the NMDA receptors; (<b>A</b>) Graphical representation of the subunit composition of NMDA receptors. Each monomer of the receptor contains four functional domains, the amino-terminal domain (ATD), ligand-binding domain (LBD), transmembrane domain (TMD), and C-terminal domain. (<b>B</b>) Ion channel activity of AMPA and Kainate receptors (members of iGluR family). (<b>C</b>) Crystal structure displaying 3D conformation of the heterotetramer NMDA receptor containing GluN1 and GluN2B subunits (PDB: 6WHX); GluN3 subunits also form functional receptors with GluN1 subunit. (<b>D</b>) The proposed GluD2 ion channel activity mechanism is through interaction with pre-synaptic linker proteins; the representation is adapted from Carillo et al. SciAdv, 2021 [<a href="#B12-ijms-25-12366" class="html-bibr">12</a>].</p>
Full article ">Figure 2
<p>Activation of the “coincidence receptor”. (<b>A</b>) Initial interaction of AMPAR (blue) and NMDAR (Purple) with pre-synaptic glutamate; the receptors are inactive at the resting membrane potential (−70 mV); upon interaction with the presynaptic glutamate, NMDAR remains inactive still since Mg<sup>2+</sup> is blocking the ion channel pore, while AMPA receptor allows in-flow of Na<sup>+</sup> ions initiating the depolarization of the postsynaptic membrane. (<b>B</b>) Depolarization of the postsynaptic membrane facilitates the release of Mg<sup>2+</sup> ions from the NMAR ion channel pore, activating the receptor. (<b>C</b>) Activation of NMDAR and repetitive presynaptic glutamate release leads to increased in-flow of Ca<sup>2+</sup> and Na<sup>+</sup> ions into the postsynaptic neurons. The representation is adapted from Sprengel et al. 2022 [<a href="#B43-ijms-25-12366" class="html-bibr">43</a>]. This fundamental characteristic of NMDA receptors is disrupted in NDDs, where disease-associated variants are distributed across various domains of the GRIN proteins within NMDARs, impacting multiple physiological properties and leading to either receptor hypofunction or hyperfunction. Missense variants within the transmembrane helix may alter NMDAR surface expression and modify receptor sensitivity to endogenous agonists and inhibitors [<a href="#B2-ijms-25-12366" class="html-bibr">2</a>]. Likewise, variants in the extracellular ATD and LBD regions are often linked to receptor loss of function in DD/ID patients [<a href="#B27-ijms-25-12366" class="html-bibr">27</a>]. These alterations impact the neurons’ downstream calcium signaling, which affects long-term potentiation and synaptic plasticity in NDD patients.</p>
Full article ">Figure 3
<p>Schematics of the eight functional isoforms of the GluN1 subunit resulting from alternative RNA splicing. Color coding represents the splicing sites at exons 5, 21, and 22 or 22’, denoted as cassettes N1, C1, C2, and C2’, respectively. N1 cassette site is in the amino-terminal domain, whereas C1, C2, and C2’ sites are in the C-terminal domain. The schematics are adapted from Li et al., PNAS, 2021 [<a href="#B45-ijms-25-12366" class="html-bibr">45</a>].</p>
Full article ">Figure 4
<p>Modeling neurodevelopmental disorders using patient-derived iPSCs. The illustration depicts the general protocol for modelling human neurodevelopmental disorders (NDDs) using patient-derived induced pluripotent stem cells (iPSCs). The iPSCs can be derived from the fibroblasts, PBMCs, or lymphoblastoid cell lines (LCLs) of the patients as well as the healthy subjects (Isogenic controls). These iPSCs provide a platform to study the disease biology and explore novel therapeutic strategies through various techniques that can lead to the development of personalized and more effective treatment options for patients suffering with different NDDs. The schematics of the figure are adapted from Liu et al., Development, 2018 [<a href="#B148-ijms-25-12366" class="html-bibr">148</a>]. iPSC-derived neurons from ASD patients frequently exhibit reduced synaptic activity and altered excitatory/inhibitory signaling balance, both of which are critical to understanding synaptic dysfunctions characteristic of ASD [<a href="#B149-ijms-25-12366" class="html-bibr">149</a>,<a href="#B150-ijms-25-12366" class="html-bibr">150</a>,<a href="#B151-ijms-25-12366" class="html-bibr">151</a>,<a href="#B152-ijms-25-12366" class="html-bibr">152</a>]. With their ability to model complex synaptic processes, aberrant connectivity, and neurotransmitter imbalances, hiPSC-derived neurons serve as invaluable tools for dissecting the molecular mechanisms underlying neuropsychiatric disorders such as ASD, schizophrenia, bipolar disorder, and ID [<a href="#B153-ijms-25-12366" class="html-bibr">153</a>,<a href="#B154-ijms-25-12366" class="html-bibr">154</a>,<a href="#B155-ijms-25-12366" class="html-bibr">155</a>,<a href="#B156-ijms-25-12366" class="html-bibr">156</a>,<a href="#B157-ijms-25-12366" class="html-bibr">157</a>]. Integration-free methods for iPSC generation avoids genomic integration of vectors, thereby preserving genetic integrity and reducing tumorigenic risks [<a href="#B158-ijms-25-12366" class="html-bibr">158</a>]. Consequently, several groups have successfully adopted this method for modeling different neurological disorders. For instance, iPSCs were generated by reprogramming fibroblasts derived from a Phelan-McDermid syndrome (PMS) patient, harboring an insertion mutation in SHANK3 (C.3679insG) [<a href="#B159-ijms-25-12366" class="html-bibr">159</a>]. The iPSCs were observed to express the pluripotency markers, differentiate into the three germ layers, retain the disease-causing mutation, and display normal karyotypes. Therefore, this technology allows researchers to explore the functional properties of cellular factors involved in the pathology of NDDs, which can be translated into a patient specific therapeutic intervention.</p>
Full article ">
49 pages, 1542 KiB  
Review
From Fundamentals to Innovation in Alzheimer’s Disease: Molecular Findings and Revolutionary Therapies
by Mădălina Georgeta Sighencea, Ramona Ștefania Popescu and Simona Corina Trifu
Int. J. Mol. Sci. 2024, 25(22), 12311; https://doi.org/10.3390/ijms252212311 - 16 Nov 2024
Viewed by 1458
Abstract
Alzheimer’s disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying [...] Read more.
Alzheimer’s disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut–brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy. Full article
Show Figures

Figure 1

Figure 1
<p>The molecular mechanisms in Alzheimer’s disease pathogenesis.</p>
Full article ">Figure 2
<p>The effects of astrocytes on amyloid-beta deposition, dendrites, synapses, and mitochondria in Alzheimer’s disease. PKC: protein kinase C; PI3Ks: phosphoinositide 3-kinases; NGF: Neuron Growth Factor; BDNF: brain-derived neurotrophic factor; TGF-β: Tumor Beta Growth Factor; APP: amyloid precursor protein; IFN-γ: interferon-gamma; IL-1β: interleukin-1-beta; TNFα: Tumor Necrosis Factor alpha; Aβ: amyloid-beta; MMP-2; MMP-9: matrix metalloproteinases 2 and 9; ECE1; ECE2: endothelin converting enzymes 1 and 2; IDE: insulin-degrading enzyme; NEP: neprilysin; MAO-B: monoamine oxidase B, (+): stimulation, (-): inhibition.</p>
Full article ">Figure 3
<p>The overlap mechanisms between AD and PD.</p>
Full article ">
Back to TopTop