human umbilical cord blood
Recently Published Documents


TOTAL DOCUMENTS

1239
(FIVE YEARS 166)

H-INDEX

76
(FIVE YEARS 6)

2022 ◽  
Vol 10 (A) ◽  
pp. 6-11
Author(s):  
Yan Wisnu Prajoko ◽  
Agung Putra ◽  
Bayu Tirta Dirja ◽  
Adi Muradi Muhar ◽  
Nur Dina Amalina

BACKGROUND: Mesenchymal stem cells (MSCs) have potent immunosuppressive properties to control systemic lupus erythematosus (SLE) disease by releasing several anti-inflammatory molecules, particularly indoleamine 2, 3-dioxygenase (IDO), and increasing regulatory T cells (Treg) to control innate and adaptive immune cells. However, how MSCs release IDO to modulate Treg in controlling B is poorly understood. Therefore, investigating IDO, Treg, and B cells following MSC administration in SLE is needed. AIM: This study aimed to investigate the ameliorating effects of MSCs in controlling B cells mediated by an increase of IDO-induced Treg in PBMC of SLE patients. METHODS: This study used a post-test control group design. MSCs were obtained from human umbilical cord blood and characterized according to their surface antigen expression and multilineage differentiation capacities. PBMCs isolated from SLE patients were divided into five groups: Sham (placebo group), control, and three treatment groups. The treatment groups were treated by coculturing MSCs to PBMCs with a ratio of 1:10, 1:25, and 1:40 for 72 h incubation. Treg and B-cell levels were analyzed by flow cytometry with cytometric bead array (CBA) while the IDO levels were determined by ELISA. RESULTS: This study showed that the percentages of B cells decreased significantly in groups treated by dose-dependent MSCs, particularly in T1 and T2 groups followed by increased Treg cell percentages. These findings were aligned with the significant increase of the IDO levels. CONCLUSIONS: MSCs regulated B cells through an increase of IDO-induced Treg in SLE patients’ PBMC.


2021 ◽  
Vol 2021 ◽  
pp. 1-14
Author(s):  
Yufeng Zhang ◽  
Huaxin Deng ◽  
Zhiqiang Yang ◽  
Zhe Chen ◽  
Sheng Zhang ◽  
...  

Radiation-induced bone injury (RIBI) is one of the complications after radiotherapy for malignant tumors. However, there are no effective measures for the treatment of RIBI in clinical practice, and the mechanism of RIBI is unclear. We use a single high-dose ionizing radiation (6Gy) to analyze the effect of radiotherapy on osteoblast function. Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) were cocultured with irradiated osteoblasts to examine their therapeutic effects and mechanisms on osteoblast injury. The hUCB-MSC transplantation mouse model is used to confirm the in vivo role of hUCB-MSC treatment in radiation bone injury. Western blot analysis, qRT-PCR, immunohistochemistry, and immunofluorescence staining were used to analyze gene expression and angiogenesis. The apoptosis and migration of osteoblasts were measured by Hoechst staining, scratch test, and transwell. The differentiation of osteoblasts was measured by ALP and Alizarin red staining and transmission electron microscopy. The bone-related parameters of mice were evaluated by micro-CT analysis. We found that radiation can damage the DNA of osteoblasts; induce apoptosis; reduce the differentiation, migration, and adhesion of osteoblasts, leading to lipogenesis of bone marrow mesenchymal stem cells (BMSCs) and reducing the source of osteoblasts; and increase the number of osteoclasts in bone tissue, while MSC treatment prevents these changes. Our results reveal the inhibitory effect of radiation on osteoblast function. hUCB-MSCs can be used as a therapeutic target for the development of new therapeutic strategies for radiotherapy of bone injury diseases.


2021 ◽  
Vol 9 (A) ◽  
pp. 1245-1251
Author(s):  
Sastia Winda Astuti ◽  
Isabella Kurnia Liem ◽  
Yetty Ramli

BACKGROUND: Stroke is the second most cause of death in the world. There are several treatments but they often end up with disabilities. Recently, cell therapy has become a new hope as an alternative treatment as it could improve the patients neurological deficits and daily living activities. Cord blood mononuclear cells (CB-MNCs) are one of the cell therapies for post-ischemic neurogenesis by intravenous or intra-arterial administration; however, it is not clear which one is better. AIM: This study aims to compare the effects of intra-arterial and intravenous administration of human CB-MNC on cortical neurogenesis of rat brain after ischemic stroke. METHODS: Twenty-four rats were divided into four groups, that is, control, middle cerebral artery obstruction (MCAO) without treatment, MCAO with intra-arterial CB-MNC injection (MCAO-IA), and MCAO with intravenous CB-MNC injection (MCAO-IV). Two weeks after injection, all rats were sacrificed, the brain was harvested, histologically process and stained with hematoxylin eosin (HE) to determine cellular and tissue morphology changes, and immunohistochemical staining, anti-NeuN antibody to determine the number of cortical neurons. The HE showed that MCAO rat brain had gliosis and shrunken cells. RESULTS: The results showed that MCAO-IA and MCAO-IV had fewer areas of gliosis and shrunken cells when compared to the MCAO group. The number of neurons also showed an increase. However, there was no difference between the MCAO-IA and MCAO-IV groups. It was concluded both of them could improve neurogenesis. CONCLUSION: CB-MNC administration can be an alternative for stroke ischemic therapy because it is proven to increase neurogenesis and reduce gliosis areas. However, there was no difference in neurogenesis in the brain tissue of mice injected with CB-MNC intravenously or intra-arterially.


2021 ◽  
Vol 118 (47) ◽  
pp. e2109548118
Author(s):  
Jin Kyeong Choi ◽  
Cheng-Rong Yu ◽  
So Jin Bing ◽  
Yingyos Jittayasothorn ◽  
Mary J. Mattapallil ◽  
...  

Regulatory B cells (Breg cells) that secrete IL-10 or IL-35 (i35-Breg) play key roles in regulating immunity in tumor microenvironment or during autoimmune and infectious diseases. Thus, loss of Breg function is implicated in development of autoimmune diseases while aberrant elevation of Breg prevents sterilizing immunity, exacerbates infectious diseases, and promotes cancer metastasis. Breg cells identified thus far are largely antigen-specific and derive mainly from B2-lymphocyte lineage. Here, we describe an innate-like IL-27–producing natural regulatory B-1a cell (i27-Breg) in peritoneal cavity and human umbilical cord blood. i27-Bregs accumulate in CNS and lymphoid tissues during neuroinflammation and confers protection against CNS autoimmune disease. i27-Breg immunotherapy ameliorated encephalomyelitis and uveitis through up-regulation of inhibitory receptors (Lag3, PD-1), suppression of Th17/Th1 responses, and propagating inhibitory signals that convert conventional B cells to regulatory lymphocytes that secrete IL-10 and/or IL-35 in eye, brain, or spinal cord. Furthermore, i27-Breg proliferates in vivo and sustains IL-27 secretion in CNS and lymphoid tissues, a therapeutic advantage over administering biologics (IL-10, IL-35) that are rapidly cleared in vivo. Mutant mice lacking irf4 in B cells exhibit exaggerated increase of i27-Bregs with few i35-Bregs, while mice with loss of irf8 in B cells have abundance of i35-Bregs but defective in generating i27-Bregs, identifying IRF8/BATF and IRF4/BATF axis in skewing B cell differentiation toward i27-Breg and i35-Breg developmental programs, respectively. Consistent with its developmental origin, disease suppression by innate i27-Bregs is neither antigen-specific nor disease-specific, suggesting that i27-Breg would be effective immunotherapy for a wide spectrum of autoimmune diseases.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yu-Jen Huang ◽  
Jerry Cao ◽  
Chih-Yuan Lee ◽  
Yao-Ming Wu

Abstract Background Cirrhosis is a chronic liver disease whereby scar tissue replaces healthy liver parenchyma, leading to disruption of the liver architecture and hepatic dysfunction. Currently, there is no effective disease-modifying therapy for liver fibrosis. Recently, our group demonstrated that human umbilical cord blood (UCB) plasma possesses therapeutic effects in a rat model of acute liver failure. Methods In the current study, we tested whether exosomes (Exo) existed in UCB plasma and if they produced any antifibrotic benefits in a liver fibrosis model. Results Our results showed that UCB-Exo improved liver function and increased matrix metalloproteinase/tissue inhibitor of metalloproteinase degradation to reduce the degree of fibrosis. Moreover, UCB-Exo were found to suppress hepatic stellate cell (HSC) activity in vitro. These effects were associated with suppression of transforming growth factor-β/inhibitor of DNA binding 1 signaling. Conclusions These results further support that UCB-Exo have antifibrotic effects in mice with liver fibrosis and activated HSCs and may herald a new cell-free antifibrotic therapy.


Sign in / Sign up

Export Citation Format

Share Document