scholarly journals Elevated In Vitro Kinase Activity in Peripheral Blood Mononuclear Cells of Leucine‐Rich Repeat Kinase 2 G2019S Carriers: A Novel Enzyme‐Linked Immunosorbent Assay–Based Method

2020 ◽  
Vol 35 (11) ◽  
pp. 2095-2100 ◽  
Author(s):  
Katerina Melachroinou ◽  
Min Suk Kang ◽  
Christopher Liong ◽  
Sushma Narayan ◽  
Najah Levers ◽  
...  
2021 ◽  
Author(s):  
Jiraporn Kantapan ◽  
Nampeung Anukul ◽  
Nipapan Leetrakool ◽  
Gwenaël Rolin ◽  
Jackie Vergote ◽  
...  

Abstract Background: Cells-based therapy is a highly promising treatment paradigm in ischemic disease due to its ability to repair tissues when implanted into a damaged site. These therapeutic effects have been involving a strong paracrine component resulting from the high levels of bioactive molecules they secrete in response to the local microenvironment. Therefore, the secreted therapeutic can be modulated by preconditioning the cells during in vitro culture. Herein, we investigated the potential use of magnetic resonance imaging (MRI) probes “Iron-Quercetin complex” or IronQ for preconditioning peripheral blood mononuclear cells (PBMCs) to expand proangiogenic cells and enhance their secreted therapeutic factors. Methods: PBMCs obtained from healthy donor blood were cultured in the presence of the Iron-Quercetin complex. Preconditioning-PBMCs differentiated cells were characterized by immunostaining. An enzyme-linked immunosorbent assay to describe the secreted cytokines. In vitro migration and tubular formation using human umbilical vein endothelial cells (HUVECs) were completed to investigate the proangiogenic efficacy.Results: IronQ significantly increased mononuclear progenitor cells' proliferation and differentiation into the spindle-shape-like cells, expressing both hematopoietic and stromal cell markers. The expansion increased the number of colony-forming units (CFU-Hill). The conditioned medium obtained from IronQ-treated PBMCs contained a high level of Interleukin (IL)-8, IL-10, urokinase-type-plasminogen-activator (uPA), matrix metalloproteinases-9 (MMP-9), and tumor necrosis factor-alpha (TNF-α), and augmented migration and capillary network formation of HUVEC and fibroblast cells in vitro.Conclusions: Our study demonstrated that the IronQ-precondition PBMCs protocol could enhance the angiogenic and reparative potential of non-mobilized PBMCs. This protocol can be used as an adjunctive strategy to improve cell therapy's efficacy of PBMCs for ischemic diseases and chronic wound.


2021 ◽  
Vol 22 (16) ◽  
pp. 8851
Author(s):  
Jiraporn Kantapan ◽  
Nampeung Anukul ◽  
Nipapan Leetrakool ◽  
Gwenaël Rolin ◽  
Jackie Vergote ◽  
...  

Cell-based therapy is a highly promising treatment paradigm in ischemic disease due to its ability to repair tissue when implanted into a damaged site. These therapeutic effects involve a strong paracrine component resulting from the high levels of bioactive molecules secreted in response to the local microenvironment. Therefore, the secreted therapeutic can be modulated by preconditioning the cells during in vitro culturing. Herein, we investigated the potential use of magnetic resonance imaging (MRI) probes, the “iron–quercetin complex” or IronQ, for preconditioning peripheral blood mononuclear cells (PBMCs) to expand proangiogenic cells and enhance their secreted therapeutic factors. PBMCs obtained from healthy donor blood were cultured in the presence of the iron–quercetin complex. Differentiated preconditioning PBMCs were characterized by immunostaining. An enzyme-linked immunosorbent assay was carried out to describe the secreted cytokines. In vitro migration and tubular formation using human umbilical vein endothelial cells (HUVECs) were completed to investigate the proangiogenic efficacy. IronQ significantly increased mononuclear progenitor cell proliferation and differentiation into spindle-shape-like cells, expressing both hematopoietic and stromal cell markers. The expansion increased the number of colony-forming units (CFU-Hill). The conditioned medium obtained from IronQ-treated PBMCs contained high levels of interleukin 8 (IL-8), IL-10, urokinase-type-plasminogen-activator (uPA), matrix metalloproteinases-9 (MMP-9), and tumor necrosis factor-alpha (TNF-α), as well as augmented migration and capillary network formation of HUVECs and fibroblast cells, in vitro. Our study demonstrated that the IronQ-preconditioning PBMC protocol could enhance the angiogenic and reparative potential of non-mobilized PBMCs. This protocol might be used as an adjunctive strategy to improve the efficacy of cell therapy when using PBMCs for ischemic diseases and chronic wounds. However, in vivo assessment is required for further validation.


Diagnostics ◽  
2021 ◽  
Vol 11 (6) ◽  
pp. 1037
Author(s):  
Patricia Ruiz-Limon ◽  
Maria L. Ladehesa-Pineda ◽  
Clementina Lopez-Medina ◽  
Chary Lopez-Pedrera ◽  
Maria C. Abalos-Aguilera ◽  
...  

Endothelial dysfunction (ED) is well known as a process that can lead to atherosclerosis and is frequently presented in radiographic axial spondyloarthritis (r-axSpA) patients. Here, we investigated cellular and molecular mechanisms underlying r-axSpA-related ED, and analyzed the potential effect of peripheral blood mononuclear cells (PBMCs) in promoting endothelial injury in r-axSpA. A total of 30 r-axSpA patients and 32 healthy donors (HDs) were evaluated. The endothelial function, inflammatory and atherogenic profile, and oxidative stress were quantified. In vitro studies were designed to evaluate the effect of PBMCs from r-axSpA patients on aberrant endothelial activation. Compared to HDs, our study found that, associated with ED and the plasma proatherogenic profile present in r-axSpA, PBMCs from these patients displayed a pro-oxidative, proinflammatory, and proatherogenic phenotype, with most molecular changes noticed in lymphocytes. Correlation studies revealed the relationship between this phenotype and the microvascular function. Additional in vitro studies confirmed that PBMCs from r-axSpA patients promoted endothelial injury. Altogether, this study suggests the relevance of r-axSpA itself as a strong and independent cardiovascular risk factor, contributing to a dysfunctional endothelium and atherogenic status by aberrant activation of PBMCs. Lymphocytes could be the main contributors in the development of ED and subsequent atherosclerosis in this pathology.


2021 ◽  
Vol 134 ◽  
pp. 58-63
Author(s):  
Matheus Fujimura Soares ◽  
Larissa Martins Melo ◽  
Jaqueline Poleto Bragato ◽  
Amanda de Oliveira Furlan ◽  
Natália Francisco Scaramele ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document