Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (60)

Search Parameters:
Keywords = CD146 pericyte

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 11818 KiB  
Article
Cryopreservation and Validation of Microfragmented Adipose Tissue for Autologous Use in Knee Osteoarthritis Treatment
by Marija Zekušić, Petar Brlek, Lucija Zenić, Vilim Molnar, Maja Ledinski, Marina Bujić Mihica, Adela Štimac, Beata Halassy, Snježana Ramić, Dominik Puljić, Tiha Vučemilo, Carlo Tremolada, Srećko Sabalić, David C. Karli, Dimitrios Tsoukas and Dragan Primorac
Int. J. Mol. Sci. 2025, 26(14), 6969; https://doi.org/10.3390/ijms26146969 - 20 Jul 2025
Viewed by 441
Abstract
Micro-fragmented adipose tissue (MFAT) is a promising autologous therapy for knee osteoarthritis. To avoid repeated liposuction procedures for its clinical application, MFAT obtained from patients with knee osteoarthritis was stored at −80 °C in a tissue bank. This study describes the preparation, cryopreservation, [...] Read more.
Micro-fragmented adipose tissue (MFAT) is a promising autologous therapy for knee osteoarthritis. To avoid repeated liposuction procedures for its clinical application, MFAT obtained from patients with knee osteoarthritis was stored at −80 °C in a tissue bank. This study describes the preparation, cryopreservation, thawing, and washing, as well as comprehensive analysis of cell populations in fresh and MFAT thawed after two years. Immunophenotyping of both fresh and thawed MFAT showed a significant presence of endothelial progenitors and pericytes in the stromal vascular fraction. Viability before (59.75%) and after freezing (55.73%) showed no significant difference. However, the average cell count per gram of MFAT was significantly reduced in thawed samples (3.00 × 105) compared to fresh ones (5.64 × 105), likely due to processing steps. Thawed MFAT samples showed increased CD73 expression on the CD31highCD34high subset of EP and SA-ASC, as well as increased expression of CD105 on EP, the CD31lowCD34low subset of EP, pericytes, and SA-ASC. Microbiological testing confirmed 100% sterility, and double washing efficiently removed DMSO, confirming sample safety. Histological analysis revealed healthy, uniformly shaped adipocytes with intact membranes. This approach allows accurate estimation of cell yield for intra-articular injection, ensuring delivery of the target cell number into the knee. Quality control analysis confirms that cryopreserved MFAT retains high cellular and structural integrity, supporting its safety and suitability for clinical application. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

31 pages, 8559 KiB  
Article
GPX1 and RCN1 as New Endoplasmic Reticulum Stress-Related Biomarkers in Multiple Sclerosis Brain Tissue and Their Involvement in the APP-CD74 Pathway: An Integrated Study Combining Machine Learning and Multi-Omics
by Zhixin Qiao, Yanping Wang, Xiaoru Ma, Xiyu Zhang, Junfeng Wu, Anqi Li, Chao Wang, Xin Xiu, Sifan Zhang, Xiujuan Lang, Xijun Liu, Bo Sun, Hulun Li and Yumei Liu
Int. J. Mol. Sci. 2025, 26(13), 6286; https://doi.org/10.3390/ijms26136286 - 29 Jun 2025
Viewed by 689
Abstract
This study identified 13 endoplasmic reticulum stress (ERS)-related biomarkers associated with multiple sclerosis (MS) through integrated bioinformatics analysis (including weighted gene co-expression network analysis and machine learning algorithms) and single-cell sequencing, combined with validation in an experimental autoimmune encephalomyelitis (EAE) mouse model. Among [...] Read more.
This study identified 13 endoplasmic reticulum stress (ERS)-related biomarkers associated with multiple sclerosis (MS) through integrated bioinformatics analysis (including weighted gene co-expression network analysis and machine learning algorithms) and single-cell sequencing, combined with validation in an experimental autoimmune encephalomyelitis (EAE) mouse model. Among them, GPX1, RCN1, and UBE2D3 exhibited high diagnostic value (AUC > 0.7, p < 0.05), and the diagnostic potential of GPX1 and RCN1 was confirmed in the animal model. The study found that memory B cells, plasma cells, neutrophils, and M1 macrophages were significantly increased in MS patients, while naive B cells and activated NK cells decreased. Consensus clustering based on key ERS-related genes divided MS patients into two subtypes. Single-cell sequencing showed that microglia and pericytes were the cell types with the highest expression of key ERS-related genes, and the APP-CD74 pathway was enhanced in the brain tissue of MS patients. Mendelian randomization analysis suggested that GPX1 plays a protective role in MS. These findings reveal the mechanisms of ERS-related biomarkers in MS and provide potential targets for diagnosis and treatment. Full article
(This article belongs to the Special Issue Applications of Machine Learning in Bioinformatics and Biomedicine)
Show Figures

Figure 1

14 pages, 3107 KiB  
Article
The Pro-Angiogenic Potential of Periodontal Ligament Stem Cells and Dental Pulp Stem Cells: A Comparative Analysis
by Ilaria Roato, Clarissa Orrico, Sara Meinardi, Riccardo Pedraza, Alessandro Mosca Balma, Giacomo Baima, Tullio Genova, Mario Aimetti and Federico Mussano
Cells 2025, 14(12), 864; https://doi.org/10.3390/cells14120864 - 8 Jun 2025
Viewed by 562
Abstract
The role of periodontal ligament stem cells (PDLSCs) and dental pulp stem cells (DPSCs) in stimulating angiogenesis has been reported, but their angiogenetic potential has not been directly compared. In this work, paired PDLSCs and DPSCs, i.e., derived from the same donor, were [...] Read more.
The role of periodontal ligament stem cells (PDLSCs) and dental pulp stem cells (DPSCs) in stimulating angiogenesis has been reported, but their angiogenetic potential has not been directly compared. In this work, paired PDLSCs and DPSCs, i.e., derived from the same donor, were tested for their immunophenotype and multi-differentiation capabilities, with particular emphasis on their pro-angiogenic activity. Flow cytometry was utilized to study the expression of mesenchymal stem cell, pericyte, and endothelial markers, while gene expression was evaluated through real-time PCR. The angiogenic potential was assessed recurring to tubulogenesis assay, co-cultures with Human Microvascular Endothelial Cell (HMEC-1), and VEGF-A quantification. The immunophenotype of DPSCs and PDLSCs was different in CD146+ and CD31+ cell subsets, but both cell types promoted HMEC-1 tubulogenesis in vitro. Consistently, VEGF-A gene expression level and its quantification in cell-conditioned media of PDLSCs and DPSCs was comparable between them, and both promoted the formation of vessel-like structures, when co-cultured with HMEC-1 cells. All together, these results showed the heterogeneity of PDLSCs and DPSCs, which are constituted of different cellular subsets, likely modulated by the microenvironmental cues. PDLSCs and DPSCs showed comparable pro-angiogenic activity, enhanced by the contemporary expression of angiogenic and chemotactic factors. Full article
Show Figures

Graphical abstract

11 pages, 4050 KiB  
Brief Report
Spatiotemporal Angiogenic Patterns in the Development of the Mouse Fetal Blood–Brain Barrier System During Pregnancy
by Samuel Nofsinger Brown, Philemon Shallie, Connor A. Sierra, Neha Nayak, Anthony O. Odibo, Paula Monaghan-Nichols and Nihar R. Nayak
Int. J. Mol. Sci. 2025, 26(8), 3862; https://doi.org/10.3390/ijms26083862 - 18 Apr 2025
Viewed by 614
Abstract
Understanding the timing of fetal brain vulnerability to inflammatory changes in pregnancy complications is crucial for predicting neurodevelopmental risks. Beyond the placenta, the developing brain’s vascular system is believed to form a secondary defense, the blood–brain barrier (BBB), which restricts harmful substances that [...] Read more.
Understanding the timing of fetal brain vulnerability to inflammatory changes in pregnancy complications is crucial for predicting neurodevelopmental risks. Beyond the placenta, the developing brain’s vascular system is believed to form a secondary defense, the blood–brain barrier (BBB), which restricts harmful substances that could disrupt neurodevelopment. However, the precise timing and mechanisms underlying BBB development are poorly understood. In this study, we examined the spatiotemporal expression of key BBB components and fetal brain vascularization in mice from gestational days (GD) 10 to 18. Fetal brain sections were immunostained to identify BBB components, including CD31, Factor VIII, NG2, and claudin-5. Our results showed that endothelial precursor cells form the primitive vascular network in a caudal-to-rostral gradient by GD10, with pericyte recruitment stabilizing vessels by GD12 in a lateral-to-medial gradient that aligns with neurogenesis, despite some regional exceptions. However, Factor VIII was not detected until GD15, and claudin-5 until GD18, suggesting a significant delay in endothelial maturation and tight junction formation. These findings highlight the critical timing of structural developments in the fetal brain vasculature and its vulnerability to placental diseases, laying the groundwork for future research on the impact of placental disorders on fetal brain development and potential therapeutic interventions. Full article
(This article belongs to the Special Issue Molecular Regulation in Female Reproduction 2.0)
Show Figures

Figure 1

22 pages, 2952 KiB  
Article
Chondrogenic and Osteogenic In Vitro Differentiation Performance of Unsorted and Sorted CD34+, CD146+, and CD271+ Stem Cells Derived from Microfragmented Adipose Tissue of Patients with Knee Osteoarthritis
by Jasmin Bagge, Haider Mahmood, Jennifer Janes, Kilian Vomstein, Lars Blønd, Lisbet R. Hölmich, Kristine Freude, Jan O. Nehlin, Kristoffer W. Barfod and Per Hölmich
J. Clin. Med. 2025, 14(4), 1184; https://doi.org/10.3390/jcm14041184 - 11 Feb 2025
Viewed by 1170
Abstract
Background/Objectives: Treatment of knee osteoarthritis (OA) with autologous stem cells from microfragmented adipose tissue (MFAT) has shown promising but varying results. Multiple stem cell types, including CD34+, CD146+, and CD271+ stem cells, have been identified within MFAT. [...] Read more.
Background/Objectives: Treatment of knee osteoarthritis (OA) with autologous stem cells from microfragmented adipose tissue (MFAT) has shown promising but varying results. Multiple stem cell types, including CD34+, CD146+, and CD271+ stem cells, have been identified within MFAT. Patient-specific heterogeneity in stem cell populations and the content of highly potent cells may be determining factors for a successful treatment outcome. The current study aimed to identify the most promising stem cell type in MFAT to treat OA, focusing on their chondrogenic and osteogenic differentiation performance. Methods: CD34+, CD146+, and CD271+ stem cells from the MFAT of eight patients with knee OA were separated using magnetic-activated cell sorting (MACS) and analyzed as subtypes. Unsorted cells were used as a control. Chondrogenic and osteogenic in vitro differentiation were assessed through Safranin-O and H&E staining, pellet size, and qPCR for chondrogenesis, as well as Alizarin Red S staining and qPCR for osteogenesis. Results: CD34+, CD146+, and CD271+ stem cells were doubled using MACS. All subtypes were able to undergo osteogenic differentiation with Alizarin Red S staining, revealing a significant increase in calcium deposits of induced cells compared to non-induced controls. CD146+ stem cells showed higher calcium deposition compared to CD34+, CD271+, and unsorted stem cells. All cell types could form chondrogenic pellets. CD271+ stem cells produced more proteoglycans, as shown by Safranin-O staining, than CD34+ and CD146+ stem cells, but not more than the unsorted stem cells. After differentiation induction, all cell types showed an upregulation of most chondrogenic and osteogenic biomarkers. Conclusions: CD146+ stem cells showed the highest osteogenic differentiation performance for calcium deposition, while CD271+ stem cells showed the greatest chondrogenic differentiation performance for proteoglycan formation. The prevalence of these stem cell types may play a critical role in the clinical effectiveness when treating OA. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

19 pages, 9839 KiB  
Article
Expression of Lumican and Osteopontin in Perivascular Areas of the Glioblastoma Peritumoral Niche and Its Value for Prognosis
by María Dolores Salinas, Pablo Rodriguez, Gonzalo Rubio and Rut Valdor
Int. J. Mol. Sci. 2025, 26(1), 192; https://doi.org/10.3390/ijms26010192 - 29 Dec 2024
Cited by 1 | Viewed by 1272
Abstract
Glioblastoma (GB) is one of the most aggressive and treatment-resistant cancers due to its complex tumor microenvironment (TME). We previously showed that GB progression is dependent on the aberrant induction of chaperone-mediated autophagy (CMA) in pericytes (PCs), which promotes TME immunosuppression through the [...] Read more.
Glioblastoma (GB) is one of the most aggressive and treatment-resistant cancers due to its complex tumor microenvironment (TME). We previously showed that GB progression is dependent on the aberrant induction of chaperone-mediated autophagy (CMA) in pericytes (PCs), which promotes TME immunosuppression through the PC secretome. The secretion of extracellular matrix (ECM) proteins with anti-tumor (Lumican) and pro-tumoral (Osteopontin, OPN) properties was shown to be dependent on the regulation of GB-induced CMA in PCs. As biomarkers are rarely studied in TME, in this work, we aimed to validate Lumican and OPN as prognostic markers in the perivascular areas of the peritumoral niche of a cohort of GB patients. Previously, we had validated their expression in GB xenografted mice presenting GB infiltration (OPN) or GB elimination (Lumican) dependent on competent or deficient CMA PCs, respectively. Then, patient sample classification by GB infiltration into the peritumoral brain parenchyma was related to GB-induced CMA in microvasculature PCs, analyzing the expression of the lysosomal receptor, LAMP-2A. Our results revealed a correlation between GB-induced CMA activity in peritumoral PCs and GB patients’ outcomes, identifying three degrees of severity. The perivascular expression of both immune activation markers, Iba1 and CD68, was related to CMA-dependent PC immune function and determined as useful for efficient GB prognosis. Lumican expression was identified in perivascular areas of patients with less severe outcome and partially co-localizing with PCs presenting low CMA activity, while OPN was primarily found in perivascular areas of patients with poor outcome and partially co-localizing with PCs presenting high CMA activity. Importantly, we found sex differences in the incidence of middle-aged patients, being significantly higher in men but with worse prognosis in women. Our results confirmed that Lumican and OPN in perivascular areas of the GB peritumoral niche are effective predictive biomarkers for evaluating prognosis and monitoring possible therapeutic immune responses dependent on PCs in tumor progression. Full article
(This article belongs to the Special Issue New Wave of Cancer Therapeutics: Challenges and Opportunities)
Show Figures

Figure 1

15 pages, 37035 KiB  
Article
Platelet-Derived Growth Factor Subunit A Strengthens the Neurovascular Unit and Inhibits Retinal Vascular Regression Under Hyperoxic Conditions
by Kaito Yokota, Haruhiko Yamada, Hidetsugu Mori, Yuki Hattori, Masatoshi Omi, Yuichi Yamamoto, Keiko Toyama and Hisanori Imai
Int. J. Mol. Sci. 2024, 25(23), 12945; https://doi.org/10.3390/ijms252312945 - 2 Dec 2024
Cited by 1 | Viewed by 1091
Abstract
Retinopathy of prematurity (ROP) is primarily caused by the exposure of preterm infants with underdeveloped blood vessels to high oxygen concentrations. This damages the astrocytes that promote normal vascular development, leading to avascularity, pathological neovascularization, and retinal detachment, and even blindness as the [...] Read more.
Retinopathy of prematurity (ROP) is primarily caused by the exposure of preterm infants with underdeveloped blood vessels to high oxygen concentrations. This damages the astrocytes that promote normal vascular development, leading to avascularity, pathological neovascularization, and retinal detachment, and even blindness as the disease progresses. In this study, the aim was to investigate the differences in the characteristics of astrocytes and blood vessels between wild-type (WT) and genetically modified mice overexpressing platelet-derived growth factor subunit A (PDGF-A) in the retina immediately after high oxygen exposure, a protocol in the oxygen-induced retinopathy (OIR) model of ROP. Our results showed that PDGF-A mice exhibited an increased population of astrocytes and higher vascular density than WT mice and that PDGF-A strengthened the resistance to hyperoxic conditions. In the OIR model, PDGF-A mice had reduced avascular zone areas following hyperoxia exposure. Furthermore, immunostaining for NG2 and CD31 showed that pericytes tended to regress earlier than endothelial cells, particularly at the vessel edges in both WT and transgenic mice, indicating relatively higher susceptibility to hyperoxia-induced damage. These findings suggest that PDGF-A plays a crucial role in stabilizing retinal vessels and may serve as a novel therapeutic target for ROP, highlighting the potential significance of PDGF-A in the pathological mechanisms of retinal diseases. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

18 pages, 7398 KiB  
Article
L-PGDS–PGD2–DP1 Axis Regulates Phagocytosis by CD36+ MGs/MΦs That Are Exclusively Present Within Ischemic Areas After Stroke
by Takayuki Nakagomi, Aya Narita, Hideaki Nishie, Akiko Nakano-Doi, Toshinori Sawano, Yu Fukuda and Tomohiro Matsuyama
Cells 2024, 13(20), 1737; https://doi.org/10.3390/cells13201737 - 20 Oct 2024
Cited by 3 | Viewed by 2012
Abstract
Brain injuries, such as ischemic stroke, cause cell death. Although phagocytosis of cellular debris is mainly performed by microglia/macrophages (MGs/MΦs), excessive accumulation beyond their phagocytic capacities results in waste product buildup, delaying brain cell regeneration. Therefore, it is essential to increase the potential [...] Read more.
Brain injuries, such as ischemic stroke, cause cell death. Although phagocytosis of cellular debris is mainly performed by microglia/macrophages (MGs/MΦs), excessive accumulation beyond their phagocytic capacities results in waste product buildup, delaying brain cell regeneration. Therefore, it is essential to increase the potential for waste product removal from damaged brains. Lipocalin-type prostaglandin D synthase (L-PGDS) is the primary synthase for prostaglandin D2 (PGD2) and has been reported as a scavenger of waste products. However, the mechanism by which the L-PGDS–PGD2 axis exerts such an effect remains unelucidated. In this study, using a mouse model of ischemic stroke, we found that L-PGDS and its downstream signaling pathway components, including PGD2 and PGD2 receptor DP1 (but not DP2), were significantly upregulated in ischemic areas. Immunohistochemistry revealed the predominant expression of L-PGDS in the leptomeninges of ischemic areas and high expression levels of DP1 in CD36+ MGs/MΦs that were specifically present within ischemic areas. Furthermore, PGD2 treatment promoted the conversion of MGs/MΦs into CD36+ scavenger types and increased phagocytic activities of CD36+ MGs/MΦs. Because CD36+ MGs/MΦs specifically appeared within ischemic areas after stroke, our findings suggest that the L-PGDS–PGD2–DP1 axis plays an important role in brain tissue repair by regulating phagocytic activities of CD36+ MGs/MΦs. Full article
(This article belongs to the Special Issue New Advances in Neuroinflammation)
Show Figures

Graphical abstract

11 pages, 4198 KiB  
Article
Increasing Endoglin Deletion in Endothelial Cells Exacerbates the Severity of Brain Arteriovenous Malformation in Mouse
by Zahra Shabani, Leandro Barbosa Do Prado, Rui Zhang, Wan Zhu, Sonali S. Shaligram, Alka Yadav, Calvin Wang and Hua Su
Biomedicines 2024, 12(8), 1691; https://doi.org/10.3390/biomedicines12081691 - 30 Jul 2024
Cited by 1 | Viewed by 2986
Abstract
Endoglin (ENG) mutation causes type 1 hereditary hemorrhagic telangiectasia (HHT1). HHT1 patients have arteriovenous malformations (AVMs) in multiple organs, including the brain. In mice, Eng deletion induced by R26RCreER or SM22αCre leads to AVM development in the brain and other organs. [...] Read more.
Endoglin (ENG) mutation causes type 1 hereditary hemorrhagic telangiectasia (HHT1). HHT1 patients have arteriovenous malformations (AVMs) in multiple organs, including the brain. In mice, Eng deletion induced by R26RCreER or SM22αCre leads to AVM development in the brain and other organs. We hypothesized that an increase in Eng- negative ECs will enhance AVM severity. To increase EC Eng deletion, we used a codon-improved cre (icre), which is more potent in recombination of the floxed alleles than the wild-type (WT) cre. R26RCreER;Engf/f mice that have a Rosa promoter driving and tamoxifen (TM)-inducible WT cre expression globally, and PdgfbiCreER;Engf/f mice that have a Pdgfb promoter driving and TM-inducible icre expression in ECs were treated with three intra-peritoneal injections of TM (2.5 mg/25 g of body weight) to delete Eng globally or in the ECs. AAV-VEGF was stereotactically injected into the brain to induce brain focal angiogenesis and brain AVM. We found that icre caused more Eng deletion in the brain, indicated by a lower level of Eng proteins (p < 0.001) and fewer Eng-positive ECs (p = 0.01) than mice with WT cre. Mice with icre-mediated Eng deletion have more abnormal vessels (p = 0.02), CD68+ macrophages (p = 0.002), and hemorrhage (p = 0.04) and less vascular pericyte and smooth muscle coverage than mice with WT cre. In addition, arteriovenous shunts were detected in the intestines of icre mice, a phenotype that has not been detected in WT cre mice before. RNA-seq analysis showed that 8 out of the 10 top upregulated pathways identified by gene ontology (GO) analysis are related to inflammation. Therefore, the increase in Eng deletion in ECs exacerbates AVM severity, which is associated with enhanced inflammation. Strategies that can reduce Eng-negative ECs could be used to develop new therapies to reduce AVM severity for HHT1 patients. Full article
Show Figures

Figure 1

14 pages, 2873 KiB  
Article
Chemical Coaxing of Mesenchymal Stromal Cells by Drug Repositioning for Nestin Induction
by Sun-Ung Lim, Dae-Won Lee, Jung-Ho Kim, Young-Ju Kang, In-Yong Kim and Il-Hoan Oh
Int. J. Mol. Sci. 2024, 25(15), 8006; https://doi.org/10.3390/ijms25158006 - 23 Jul 2024
Viewed by 1432
Abstract
Mesenchymal stromal cells (MSCs) display heterogeneity in origin and functional role in tissue homeostasis. Subsets of MSCs derived from the neural crest express nestin and serve as niches in bone marrow, but the possibility of coaxing MSCs into nestin-expresing cells for enhanced supportive [...] Read more.
Mesenchymal stromal cells (MSCs) display heterogeneity in origin and functional role in tissue homeostasis. Subsets of MSCs derived from the neural crest express nestin and serve as niches in bone marrow, but the possibility of coaxing MSCs into nestin-expresing cells for enhanced supportive activity is unclear. In this study, as an approach to the chemical coaxing of MSC functions, we screened libraries of clinically approved chemicals to identify compounds capable of inducing nestin expression in MSCs. Out of 2000 clinical compounds, we chose vorinostat as a candidate to coax the MSCs into neural crest-like fates. When treated with vorinostat, MSCs exhibited a significant increase in the expression of genes involved in the pluripotency and epithelial–mesenchymal transition (EMT), as well as nestin and CD146, the markers for pericytes. In addition, these nestin-induced MSCs exhibited enhanced differentiation towards neuronal cells with the upregulation of neurogenic markers, including SRY-box transcription factor 2 (Sox2), SRY-box transcription factor 10 (Sox10) and microtubule associated protein 2 (Map2) in addition to nestin. Moreover, the coaxed MSCs exhibited enhanced supporting activity for hematopoietic progenitors without supporting leukemia cells. These results demonstrate the feasibility of the drug repositioning of MSCs to induce neural crest-like properties through the chemical coaxing of cell fates. Full article
Show Figures

Figure 1

14 pages, 2709 KiB  
Article
Therapeutic Anti-Tumor Efficacy of DC-Based Vaccines Targeting TME-Associated Antigens Is Improved When Combined with a Chemokine-Modulating Regimen and/or Anti-PD-L1
by Jennifer L. Taylor, Kathleen M. Kokolus, Per H. Basse, Jessica N. Filderman, Chloe E. Cosgrove, Simon C. Watkins, Andrea Gambotto, Devin B. Lowe, Robert P. Edwards, Pawel Kalinski and Walter J. Storkus
Vaccines 2024, 12(7), 777; https://doi.org/10.3390/vaccines12070777 - 15 Jul 2024
Cited by 2 | Viewed by 3348
Abstract
We previously reported that dendritic cell (DC)-based vaccines targeting antigens expressed by tumor-associated vascular endothelial cells (VECs) and pericytes effectively control tumor growth in translational mouse tumor models. In the current report, we examined whether the therapeutic benefits of such tumor blood vessel [...] Read more.
We previously reported that dendritic cell (DC)-based vaccines targeting antigens expressed by tumor-associated vascular endothelial cells (VECs) and pericytes effectively control tumor growth in translational mouse tumor models. In the current report, we examined whether the therapeutic benefits of such tumor blood vessel antigen (TBVA)-targeted vaccines could be improved by the cotargeting of tumor antigens in the s.c. B16 melanoma model. We also evaluated whether combination vaccines incorporating anti-PD-L1 checkpoint blockade and/or a chemokine-modulating (CKM; IFNα + TLR3-L [rintatolimod] + Celecoxib) regimen would improve T cell infiltration/functionality in tumors yielding enhanced treatment benefits. We report that DC–peptide or DC–tumor lysate vaccines coordinately targeting melanoma antigens and TBVAs were effective in slowing B16 growth in vivo and extending survival, with superior outcomes observed for DC–peptide-based vaccines. Peptide-based vaccines that selectively target either melanoma antigens or TBVAs elicited a CD8+ T cell repertoire recognizing both tumor cells and tumor-associated VECs and pericytes in vitro, consistent with a treatment-induced epitope spreading mechanism. Notably, combination vaccines including anti-PD-L1 + CKM yielded superior therapeutic effects on tumor growth and animal survival, in association with the potentiation of polyfunctional CD8+ T cell reactivity against both tumor cells and tumor-associated vascular cells and a pro-inflammatory TME. Full article
Show Figures

Figure 1

24 pages, 7395 KiB  
Article
Single-Cell Analyses Offer Insights into the Different Remodeling Programs of Arteries and Veins
by Miguel G. Rojas, Simone Pereira-Simon, Zachary M. Zigmond, Javier Varona Santos, Mikael Perla, Nieves Santos Falcon, Filipe F. Stoyell-Conti, Alghidak Salama, Xiaofeng Yang, Xiaochun Long, Juan C. Duque, Loay H. Salman, Marwan Tabbara, Laisel Martinez and Roberto I. Vazquez-Padron
Cells 2024, 13(10), 793; https://doi.org/10.3390/cells13100793 - 7 May 2024
Cited by 7 | Viewed by 3789
Abstract
Arteries and veins develop different types of occlusive diseases and respond differently to injury. The biological reasons for this discrepancy are not well understood, which is a limiting factor for the development of vein-targeted therapies. This study contrasts human peripheral arteries and veins [...] Read more.
Arteries and veins develop different types of occlusive diseases and respond differently to injury. The biological reasons for this discrepancy are not well understood, which is a limiting factor for the development of vein-targeted therapies. This study contrasts human peripheral arteries and veins at the single-cell level, with a focus on cell populations with remodeling potential. Upper arm arteries (brachial) and veins (basilic/cephalic) from 30 organ donors were compared using a combination of bulk and single-cell RNA sequencing, proteomics, flow cytometry, and histology. The cellular atlases of six arteries and veins demonstrated a 7.8× higher proportion of contractile smooth muscle cells (SMCs) in arteries and a trend toward more modulated SMCs. In contrast, veins showed a higher abundance of endothelial cells, pericytes, and macrophages, as well as an increasing trend in fibroblasts. Activated fibroblasts had similar proportions in both types of vessels but with significant differences in gene expression. Modulated SMCs and activated fibroblasts were characterized by the upregulation of MYH10, FN1, COL8A1, and ITGA10. Activated fibroblasts also expressed F2R, POSTN, and COMP and were confirmed by F2R/CD90 flow cytometry. Activated fibroblasts from veins were the top producers of collagens among all fibroblast populations from both types of vessels. Venous fibroblasts were also highly angiogenic, proinflammatory, and hyper-responders to reactive oxygen species. Differences in wall structure further explain the significant contribution of fibroblast populations to remodeling in veins. Fibroblasts are almost exclusively located outside the external elastic lamina in arteries, while widely distributed throughout the venous wall. In line with the above, ECM-targeted proteomics confirmed a higher abundance of fibrillar collagens in veins vs. more basement ECM components in arteries. The distinct cellular compositions and transcriptional programs of reparative populations in arteries and veins may explain differences in acute and chronic wall remodeling between vessels. This information may be relevant for the development of antistenotic therapies. Full article
Show Figures

Figure 1

20 pages, 18479 KiB  
Article
Pulmonary Effects of Traumatic Brain Injury in Mice: A Gene Set Enrichment Analysis
by Wei-Hung Chan, Shih-Ming Huang and Yi-Lin Chiu
Int. J. Mol. Sci. 2024, 25(5), 3018; https://doi.org/10.3390/ijms25053018 - 5 Mar 2024
Cited by 2 | Viewed by 2579
Abstract
Acute lung injury occurs in 20–25% of cases following traumatic brain injury (TBI). We investigated changes in lung transcriptome expression post-TBI using animal models and bioinformatics. Employing unilateral controlled cortical impact for TBI, we conducted microarray analysis after lung acquisition, followed by gene [...] Read more.
Acute lung injury occurs in 20–25% of cases following traumatic brain injury (TBI). We investigated changes in lung transcriptome expression post-TBI using animal models and bioinformatics. Employing unilateral controlled cortical impact for TBI, we conducted microarray analysis after lung acquisition, followed by gene set enrichment analysis of differentially expressed genes. Our findings indicate significant upregulation of inflammation-related genes and downregulation of nervous system genes. There was enhanced infiltration of adaptive immune cells, evidenced by positive enrichment in Lung-Th1, CD4, and CD8 T cells. Analysis using the Tabula Sapiens database revealed enrichment in lung-adventitial cells, pericytes, myofibroblasts, and fibroblasts, indicating potential effects on lung vasculature and fibrosis. Gene set enrichment analysis linked TBI to lung diseases, notably idiopathic pulmonary hypertension. A Venn diagram overlap analysis identified a common set of 20 genes, with FOSL2 showing the most significant fold change. Additionally, we observed a significant increase in ADRA1AIL6 production post-TBI using the L1000 library. Our study highlights the impact of brain trauma on lung injury, revealing crucial gene expression changes related to immune cell infiltration, cytokine production, and potential alterations in lung vasculature and fibrosis, along with a specific spectrum of disease influence. Full article
Show Figures

Figure 1

16 pages, 5355 KiB  
Article
In the Rat Hippocampus, Pilocarpine-Induced Status Epilepticus Is Associated with Reactive Glia and Concomitant Increased Expression of CD31, PDGFRβ, and Collagen IV in Endothelial Cells and Pericytes of the Blood–Brain Barrier
by Grigorios Kyriatzis, Anne Bernard, Angélique Bôle, Michel Khrestchatisky and Lotfi Ferhat
Int. J. Mol. Sci. 2024, 25(3), 1693; https://doi.org/10.3390/ijms25031693 - 30 Jan 2024
Cited by 6 | Viewed by 3026
Abstract
In humans and animal models, temporal lobe epilepsy (TLE) is associated with reorganization of hippocampal neuronal networks, gliosis, neuroinflammation, and loss of integrity of the blood–brain barrier (BBB). More than 30% of epilepsies remain intractable, and characterization of the molecular mechanisms involved in [...] Read more.
In humans and animal models, temporal lobe epilepsy (TLE) is associated with reorganization of hippocampal neuronal networks, gliosis, neuroinflammation, and loss of integrity of the blood–brain barrier (BBB). More than 30% of epilepsies remain intractable, and characterization of the molecular mechanisms involved in BBB dysfunction is essential to the identification of new therapeutic strategies. In this work, we induced status epilepticus in rats through injection of the proconvulsant drug pilocarpine, which leads to TLE. Using RT-qPCR, double immunohistochemistry, and confocal imaging, we studied the regulation of reactive glia and vascular markers at different time points of epileptogenesis (latent phase—3, 7, and 14 days; chronic phase—1 and 3 months). In the hippocampus, increased expression of mRNA encoding the glial proteins GFAP and Iba1 confirmed neuroinflammatory status. We report for the first time the concomitant induction of the specific proteins CD31, PDGFRβ, and ColIV—which peak at the same time points as inflammation—in the endothelial cells, pericytes, and basement membrane of the BBB. The altered expression of these proteins occurs early in TLE, during the latent phase, suggesting that they could be associated with the early rupture and pathogenicity of the BBB that will contribute to the chronic phase of epilepsy. Full article
Show Figures

Figure 1

11 pages, 698 KiB  
Review
The Mechanisms of Altered Blood–Brain Barrier Permeability in CD19 CAR T–Cell Recipients
by Soniya N. Pinto and Giedre Krenciute
Int. J. Mol. Sci. 2024, 25(1), 644; https://doi.org/10.3390/ijms25010644 - 4 Jan 2024
Cited by 7 | Viewed by 3393
Abstract
Cluster of differentiation 19 (CD19) chimeric antigen receptor (CAR) T cells are a highly effective immunotherapy for relapsed and refractory B-cell malignancies, but their utility can be limited by the development of immune effector cell-associated neurotoxicity syndrome (ICANS). The recent discovery of CD19 [...] Read more.
Cluster of differentiation 19 (CD19) chimeric antigen receptor (CAR) T cells are a highly effective immunotherapy for relapsed and refractory B-cell malignancies, but their utility can be limited by the development of immune effector cell-associated neurotoxicity syndrome (ICANS). The recent discovery of CD19 expression on the pericytes in the blood–brain barrier (BBB) suggests an important off-target mechanism for ICANS development. In addition, the release of systemic cytokines stimulated by the engagement of CD19 with the CAR T cells can cause endothelial activation and decreased expression of tight junction molecules, further damaging the integrity of the BBB. Once within the brain microenvironment, cytokines trigger a cytokine-specific cascade of neuroinflammatory responses, which manifest clinically as a spectrum of neurological changes. Brain imaging is frequently negative or nonspecific, and treatment involves close neurologic monitoring, supportive care, interleukin antagonists, and steroids. The goal of this review is to inform readers about the normal development and microstructure of the BBB, its unique susceptibility to CD19 CAR T cells, the role of individual cytokines on specific elements of the brain’s microstructural environment, and the clinical and imaging manifestations of ICANS. Our review will link cellular pathophysiology with the clinical and radiological manifestations of a complex clinical entity. Full article
Show Figures

Figure 1

Back to TopTop