perivascular niche
Recently Published Documents


TOTAL DOCUMENTS

120
(FIVE YEARS 55)

H-INDEX

21
(FIVE YEARS 4)

Pharmaceutics ◽  
2022 ◽  
Vol 14 (1) ◽  
pp. 116
Author(s):  
Ravena Pereira do Nascimento ◽  
Balbino Lino dos Santos ◽  
Jéssika Alves Oliveira Amparo ◽  
Janaina Ribeiro Pereira Soares ◽  
Karina Costa da Silva ◽  
...  

Glioblastomas (GBMs) are tumors that have a high ability to migrate, invade and proliferate in the healthy tissue, what greatly impairs their treatment. These characteristics are associated with the complex microenvironment, formed by the perivascular niche, which is also composed of several stromal cells including astrocytes, microglia, fibroblasts, pericytes and endothelial cells, supporting tumor progression. Further microglia and macrophages associated with GBMs infiltrate the tumor. These innate immune cells are meant to participate in tumor surveillance and eradication, but they become compromised by GBM cells and exploited in the process. In this review we discuss the context of the GBM microenvironment together with the actions of flavonoids, which have attracted scientific attention due to their pharmacological properties as possible anti-tumor agents. Flavonoids act on a variety of signaling pathways, counteracting the invasion process. Luteolin and rutin inhibit NFκB activation, reducing IL-6 production. Fisetin promotes tumor apoptosis, while inhibiting ADAM expression, reducing invasion. Naringenin reduces tumor invasion by down-regulating metalloproteinases expression. Apigenin and rutin induce apoptosis in C6 cells increasing TNFα, while decreasing IL-10 production, denoting a shift from the immunosuppressive Th2 to the Th1 profile. Overall, flavonoids should be further exploited for glioma therapy.


2021 ◽  
pp. 0271678X2110685
Author(s):  
Stephanie K Bonney ◽  
Liam T Sullivan ◽  
Timothy J Cherry ◽  
Richard Daneman ◽  
Andy Y Shih

Perivascular fibroblasts (PVFs) are recognized for their pro-fibrotic role in many central nervous system disorders. Like mural cells, PVFs surround blood vessels and express Pdgfrβ. However, these shared attributes hinder the ability to distinguish PVFs from mural cells. We used in vivo two-photon imaging and transgenic mice with PVF-targeting promoters (Col1a1 or Col1a2) to compare the structure and distribution of PVFs and mural cells in cerebral cortex of healthy, adult mice. We show that PVFs localize to all cortical penetrating arterioles and their offshoots (arteriole-capillary transition zone), as well as the main trunk of only larger ascending venules. However, the capillary zone is devoid of PVF coverage. PVFs display short-range mobility along the vessel wall and exhibit distinct structural features (flattened somata and thin ruffled processes) not seen with smooth muscle cells or pericytes. These findings clarify that PVFs and mural cells are distinct cell types coexisting in a similar perivascular niche.


Cells ◽  
2021 ◽  
Vol 10 (12) ◽  
pp. 3594
Author(s):  
Yin-Hung Chu ◽  
Wen-Chieh Liao ◽  
Ying-Jui Ho ◽  
Chih-Hsien Huang ◽  
To-Jung Tseng ◽  
...  

Chondroitin sulfate (CS) is a major component of the extracellular matrix found to be abnormally accumulated in several types of cancer tissues. Previous studies have indicated that CS synthases and modification enzymes are frequently elevated in human gliomas and are associated with poor prognosis. However, the underlying mechanisms of CS in cancer progression and approaches for interrupting its functions in cancer cells remain largely unexplored. Here, we have found that CS was significantly enriched surrounding the vasculature in a subset of glioma tissues, which was akin to the perivascular niche for cancer-initiating cells. Silencing or overexpression of the major CS synthase, chondroitin sulfate synthase 1 (CHSY1), significantly regulated the glioma cell invasive phenotypes and modulated integrin expression. Furthermore, we identified CD44 as a crucial chondroitin sulfate proteoglycan (CSPG) that was modified by CHSY1 on glioma cells, and the suppression of CS formation on CD44 by silencing the CHSY1-inhibited interaction between CD44 and integrin β1 on the adhesion complex. Moreover, we tested the CS-specific binding peptide, resulting in the suppression of glioma cell mobility in a fashion similar to that observed upon the silencing of CHSY1. In addition, the peptide demonstrated significant affinity to CD44, promoted CD44 degradation, and suppressed integrin β1 expression in glioma cells. Overall, this study proposes a potential regulatory loop between CS, CD44, and integrin β1 in glioma cells, and highlights the importance of CS in CD44 stability. Furthermore, the targeting of CS by specific binding peptides has potential as a novel therapeutic strategy for glioma.


2021 ◽  
Vol 11 (12) ◽  
pp. 1364
Author(s):  
Michael Strug ◽  
Lusine Aghajanova

The uterus is a homeostatic organ, unwavering in the setting of monthly endometrial turnover, placental invasion, and parturition. In response to ovarian steroid hormones, the endometrium autologously prepares for embryo implantation and in its absence will shed and regenerate. Dysfunctional endometrial repair and regeneration may present clinically with infertility and abnormal menses. Asherman’s syndrome is characterized by intrauterine adhesions and atrophic endometrium, which often impacts fertility. Clinical management of infertility associated with abnormal endometrium represents a significant challenge. Endometrial mesenchymal stem cells (MSC) occupy a perivascular niche and contain regenerative and immunomodulatory properties. Given these characteristics, mesenchymal stem cells of endometrial and non-endometrial origin (bone marrow, adipose, placental) have been investigated for therapeutic purposes. Local administration of human MSC in animal models of endometrial injury reduces collagen deposition, improves angiogenesis, decreases inflammation, and improves fertility. Small clinical studies of autologous MSC administration in infertile women with Asherman’s Syndrome suggested their potential to restore endometrial function as evidenced by increased endometrial thickness, decreased adhesions, and fertility. The objective of this review is to highlight translational and clinical studies investigating the use of MSC for endometrial dysfunction and infertility and to summarize the current state of the art in this promising area.


2021 ◽  
Vol 7 (45) ◽  
Author(s):  
James W. Opzoomer ◽  
Joanne E. Anstee ◽  
Isaac Dean ◽  
Emily J. Hill ◽  
Ihssane Bouybayoune ◽  
...  

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi206-vi206
Author(s):  
Angad Beniwal ◽  
Saket Jain ◽  
Sumedh Shah ◽  
Sabraj Gill ◽  
Garima Yagnik ◽  
...  

Abstract Among clinical analyses, elevated neutrophil-lymphocyte ratio has been correlated with poor outcomes of glioblastoma patients independent of other prognostic factors. Additionally, our flow cytometric studies of primary patient samples found neutrophil percentage to be significantly higher in higher-grade glioma versus lower-grade glioma. Tumor-associated neutrophils (TANs) comprise less than 2% of the glioblastoma microenvironment. While TANs were initially considered passive bystanders due to their short-lived nature, investigation of TANs in other cancers revealed distinct pro-tumoral roles. Therefore, we transcriptomically characterized glioblastoma TANs and defined their oncologic effects. Transcriptomic analysis of patient-matched TANs versus peripheral blood neutrophils revealed that functionally quiescent circulating neutrophils infiltrate IDH1-wild type glioblastoma via leukotriene B4 chemoattraction, where tumor cells morphologically and transcriptomically activate them to become TANs. Single-cell RNA-sequencing of patient-matched TANs and peripheral blood neutrophils revealed a subset of tumor-activated neutrophils which adopt a pro-tumoral secretory phenotype, marked by activation of the IL-17 signaling pathway and high osteopontin production. Using immunofluorescence stains of primary patient glioblastoma sections, we demonstrated that activated, myeloperoxidase-positive TANs reside in the perivascular niche of glioblastoma in close proximity to glioblastoma stem-like cells (GSCs) and CD31-positive endothelial cells. Further analysis in culture demonstrated that TAN-secreted osteopontin drives the formation, self-renewal, and proliferation of GSC-containing neurospheres. These results were validated using a syngeneic stem cell-derived IDH1-wild type murine glioblastoma model in vivo. Thus, while TANs are rare in glioblastoma, their enrichment in the glioblastoma perivascular niche uniquely positions them to support the GSCs that are crucial to therapeutic resistance of GBM.


Cells ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 2970
Author(s):  
Yuki Shirakawa ◽  
Kunimasa Ohta ◽  
Shunsuke Miyake ◽  
Ayumi Kanemaru ◽  
Akari Kuwano ◽  
...  

Although glioblastoma (GBM) stem-like cells (GSCs), which retain chemo-radio resistance and recurrence, are key prognostic factors in GBM patients, the molecular mechanisms of GSC development are largely unknown. Recently, several studies revealed that extrinsic ribosome incorporation into somatic cells resulted in stem cell properties and served as a key trigger and factor for the cell reprogramming process. In this study, we aimed to investigate the mechanisms underlying GSCs development by focusing on extrinsic ribosome incorporation into GBM cells. Ribosome-induced cancer cell spheroid (RICCS) formation was significantly upregulated by ribosome incorporation. RICCS showed the stem-like cell characters (number of cell spheroid, stem cell markers, and ability for trans differentiation towards adipocytes and osteocytes). In RICCS, the phosphorylation and protein expression of ribosomal protein S6 (RPS6), an intrinsic ribosomal protein, and STAT3 phosphorylation were upregulated, and involved in the regulation of cell spheroid formation. Consistent with those results, glioma-derived extrinsic ribosome also promoted GBM-RICCS formation through intrinsic RPS6 phosphorylation. Moreover, in glioma patients, RPS6 phosphorylation was dominantly observed in high-grade glioma tissues, and predominantly upregulated in GSCs niches, such as the perinecrosis niche and perivascular niche. Those results indicate the potential biological and clinical significance of extrinsic ribosomal proteins in GSC development.


2021 ◽  
Author(s):  
Yasmin Ghochani ◽  
Alireza Sohrabi ◽  
Sree Deepthi Deepthi Muthukrishnan ◽  
Riki Kawaguchi ◽  
Michael Condro ◽  
...  

Glioblastoma (GBM) is characterized by extensive microvascular hyperproliferation. In addition to supplying blood to the tumor, GBM vessels also provide trophic support to glioma cells and serve as conduits for migration into the surrounding brain promoting recurrence. Here, we enriched CD31-expressing glioma vascular cells (GVC) and A2B5-expressing glioma tumor cells (GTC) from primary GBM and utilized RNA sequencing to create a comprehensive interaction map of the secreted and extracellular factors elaborated by GVC that can interact with receptors and membrane molecules on GTC. To validate our findings, we utilized functional assays, including a novel hydrogel-based migration assay and in vivo mouse models to demonstrate that one identified factor, the little-studied integrin binding sialoprotein (IBSP) enhances tumor growth and promotes the migration of GTC along the vasculature. This perivascular niche interactome will serve a resource to the research community in defining the potential functions of the GBM vasculature.


2021 ◽  
Author(s):  
Wajd N. Al-Holou ◽  
Hanxiao Wang ◽  
Visweswaran Ravikumar ◽  
Morgan Oneka ◽  
Roel GW Verhaak ◽  
...  

AbstractGlioblastoma(GBM) is a lethal disease characterized by treatment resistance and recurrence. To investigate the mechanisms that drive treatment resistance in GBM, we developed a longitudinal in vivo recurrence model utilizing patient-derived GBM explants to produce paired specimens pre- and post-recurrence following temozolomide(TMZ) and radiation(IR) therapy. These studies revealed in replicate cohorts, a common gene expression profile upon recurrence, characterized by an upregulation of transcripts associated with a mesenchymal and stem cell phenotype, including TGFβ1, TGFβ2, SOX2, ZEB2, GLI2 and THY1(CD90), with greater than one-hundred-fold increase in THY1 levels. Analyses of clinical databases revealed the association of this transcriptional profile with worse overall survival and elevation in recurrent tumors. We then isolated THY1-positive cells from treatment-naïve patient samples which demonstrated inherent resistance to chemoradiation when implanted intracranially. Additionally, using image-guided biopsies from treatment-naïve human GBM we conducted spatial whole transcriptomic analyses. This revealed rare THY1+ regions characterized by elevation of the mesenchymal and stem-like gene expression profile, previously identified in our in vivo recurrent samples, which co-localized with a macrophage gene signature within the perivascular niche. Since TGFβ signaling contributes to a mesenchymal/stem-like phenotype and therapeutic resistance, and to investigate its effect on THY1, we inhibited TGFβRI kinase activity in vivo which resulted in decreased expression of genes characteristic of a mesenchymal/stem-like phenotype, including THY1. Notably, TGFβRI inhibition restored sensitivity to TMZ/IR in recurrent tumors in vivo. These studies reveal that post-TMZ/IR recurrence may result from tumor repopulation by pre-existing, therapy-resistant, THY1-positive mesenchymal/stem-like cells within the perivascular niche.One Sentence SummaryTHY1 positive tumor cells drive resistance in glioblastoma


2021 ◽  
Vol 3 (2) ◽  
Author(s):  
Elena N Tolkunova

  Gliomas are solid brain tumors composed of tumor cells and recruited heterogenic stromal component. The study of the interactions between the perivascular niche and its surrounding cells is of great value in unraveling mechanisms of drug resistance in malignant gliomas.   In this study, we isolated the stromal diploid cell population from oligodendroglioma and a mixed population of tumor aneuploid and stromal diploid cells from astrocytoma specimens. The stromal cells expressed neural stem/progenitor and mesenchymal markers showing the same discordant phenotype that is typical for glioma cells. Moreover, some of the stromal cells expressed CD133. For the first time, we demonstrated that this type of stromal cells had the typical myofibroblastic phenotype as the α-SMA+ cells formed α-SMA fibers and exhibited the specific function to deposit extracellular matrix (ECM) proteins at least in vitro. Immunofluorescent analysis showed diffuse or focal α-SMA staining in the cytoplasm of the astrocytoma-derived, A172, T98G, and U251MG glioma cells. We could suggest that α-SMA may be one of the main molecules, bearing protective functions. Possible mechanisms and consequences of α-SMA disruptions in gliomas are discussed.


Sign in / Sign up

Export Citation Format

Share Document