scholarly journals Senotherapeutic-like effect of Silybum marianum flower extract revealed on human skin cells

PLoS ONE ◽  
2021 ◽  
Vol 16 (12) ◽  
pp. e0260545
Author(s):  
Jieun Woo ◽  
Seoungwoo Shin ◽  
Eunae Cho ◽  
Dehun Ryu ◽  
David Garandeau ◽  
...  

Cellular senescence causes irreversible growth arrest of cells. Prolonged accumulation of senescent cells in tissues leads to increased detrimental effects due to senescence associated secretory phenotype (SASP). Recent findings suggest that elimination of senescent cells has a beneficial effect on organismal aging and lifespan. In this study, using a validated replicative senescent human dermal fibroblasts (HDFs) model, we showed that elimination of senescent cells is possible through the activation of an apoptotic mechanism. We have shown in this replicative senescence model, that cell senescence is associated with DNA damage and cell cycle arrest (p21, p53 markers). We have shown that Silybum marianum flower extract (SMFE) is a safe and selective senolytic agent targeting only senescent cells. The elimination of the cells is induced through the activation of apoptotic pathway confirmed by annexin V/propidium iodide and caspase-3/PARP staining. Moreover, SMFE suppresses the expression of SASP factors such as IL-6 and MMP-1 in senescent HDFs. In a co-culture model of senescent and young fibroblasts, we demonstrated that senescent cells impaired the proliferative capacities of young cells. Interestingly, when the co-culture is treated with SMFE, the cell proliferation rate of young cells is increased due to the decrease of the senescent burden. Moreover, we demonstrated in vitro that senescent fibroblasts trigger senescent process in normal keratinocytes through a paracrine effect. Indeed, the conditioned medium of senescent HDFs treated with SMFE reduced the level of senescence-associated beta-galactosidase (SA-β-Gal), p16INK4A and SASP factors in keratinocytes compared with CM of senescent HDFs. These results indicate that SMFE can prevent premature aging due to senescence and even reprograms aged skin. Indeed, thanks to its senolytic and senomorphic properties SMFE is a candidate for anti-senescence strategies.

Cells ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 356 ◽  
Author(s):  
Alessia Lo Curto ◽  
Simona Taverna ◽  
Maria Assunta Costa ◽  
Rosa Passantino ◽  
Giuseppa Augello ◽  
...  

Fabry disease (FD) is a lysosomal storage disorder (LSD) characterized by lysosomal accumulation of glycosphingolipids in a wide variety of cytotypes, including endothelial cells (ECs). FD patients experience a significantly reduced life expectancy compared to the general population; therefore, the association with a premature aging process would be plausible. To assess this hypothesis, miR-126-3p, a senescence-associated microRNA (SA-miRNAs), was considered as an aging biomarker. The levels of miR-126-3p contained in small extracellular vesicles (sEVs), with about 130 nm of diameter, were measured in FD patients and healthy subjects divided into age classes, in vitro, in human umbilical vein endothelial cells (HUVECs) “young” and undergoing replicative senescence, through a quantitative polymerase chain reaction (qPCR) approach. We confirmed that, in vivo, circulating miR-126 levels physiologically increase with age. In vitro, miR-126 augments in HUVECs underwent replicative senescence. We observed that FD patients are characterized by higher miR-126-3p levels in sEVs, compared to age-matched healthy subjects. We also explored, in vitro, the effect on ECs of glycosphingolipids that are typically accumulated in FD patients. We observed that FD storage substances induced in HUVECs premature senescence and increased of miR-126-3p levels. This study reinforces the hypothesis that FD may aggravate the normal aging process.


2020 ◽  
Vol 38 (11) ◽  
pp. 656-660
Author(s):  
Borislav Arabadjiev ◽  
Roumen Pankov ◽  
Ivelina Vassileva ◽  
Lyuben Sashov Petrov ◽  
Ivan Buchvarov

Pharmaceutics ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 1178
Author(s):  
Justyna Odrobińska ◽  
Magdalena Skonieczna ◽  
Dorota Neugebauer

The in vitro biochemical evaluation of the applicability of polymers carrying active substances (micelles and conjugates) was carried out. Previously designed amphiphilic graft copolymers with retinol or 4-n-butylresorcinol functionalized polymethacrylate backbone and poly(ethylene glycol) (PEG) side chains that included Janus-type heterografted copolymers containing both PEG and poly(ε-caprolactone) (PCL) side chains were applied as micellar carriers. The polymer self-assemblies were convenient to encapsulate arbutin (ARB) as the selected active substances. Moreover, the conjugates of PEG graft copolymers with ferulic acid (FA) or lipoic acid (LA) were also investigated. The permeability of released active substances through a membrane mimicking skin was evaluated by conducting transdermal tests in Franz diffusion cells. The biological response to new carriers with active substances was tested across cell lines, including normal human dermal fibroblasts (NHDF), human epidermal keratinocyte (HaCaT), as well as cancer melanoma (Me45) and metastatic human melanoma (451-Lu), for comparison. These polymer systems were safe and non-cytotoxic at the tested concentrations for healthy skin cell lines according to the MTT test. Cytometric evaluation of cell cycles as well as cell death defined by Annexin-V apoptosis assays and senescence tests showed no significant changes under action of the delivery systems, as compared to the control cells. In vitro tests confirmed the biochemical potential of these antioxidant carriers as beneficial components in cosmetic products, especially applied in the form of masks and eye pads.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2474-2474
Author(s):  
Piotr Smolewski ◽  
Agnieszka Janus ◽  
Barbara Cebula ◽  
Anna Linke ◽  
Krzysztof Jamroziak ◽  
...  

Abstract Background: Rapamycin (RAPA) is an inhibitor of mTOR kinase pathway. In vitro low doses of this agent induce cell cycle arrest in G1 phase, whereas higher concentrations of RAPA exert proapoptotic effects. Aim: We assessed cytotoxicity of RAPA alone or in combination with cytarabine (cytosine arabinoside, ARA-C) in acute myeloblastic leukemia (AML) cells and in normal lymphocytes obtained from 10 healthy volunteers. Methods: AML cells (in vitro HL-60 cell line and ex vivo leukemic cells) and phytohemaglutynin (PHA)-stimulated normal lymphocytes were treated for 24 – 48 h with 1 ng/ml RAPA alone or in combination with 50 nM cytarabine (Ara-C). Moreover, cells was pre-incubated with RAPA for 24 h and then Ara-C was added for the next 24 h. Untreated cultures and those treated with RAPA, Ara-C or PHA alone served as respective controls. The proapoptotic effect was assessed by Annexin V assay and presented as a percentage of Annexin-V-positive cells (apoptotic index; AI). Cell cycle was analyzed by DNA distribution in propydium iodide/RN-ase stained cells. Cyclin D3, A and E expression was also measured using flow cytometry. Results: Median AI induced in HL-60 cells after 24 h treatment with RAPA+Ara-C (30.1%) was significantly higher than induced by RAPA (7.2%) or Ara-C (18.5%) alone (p=0.002 and p=0.03, respectively). The RAPA+Ara-C combination exerted additive effect (combination index 0.87) in that model. Additional 24 hour pretreatment with RAPA further increased apoptosis (median AI 41.5%, vs. 10.9% after 48 h-RAPA alone). In contrast to leukemic cells, pretreatment of normal PHA-stimulated lymphocytes with RAPA caused their G1 phase cell cycle arrest, with significant decrease in cyclin D3 expression (vs. untreated cells - p<0.001). This resulted in prevention of Ara-C-induced cytotoxicity in healthy lymphocytes, when Ara-C was added for another 24 h. Importantly, that protective effect was reversible when RAPA-treated lymphocytes were rinsed and then cultured in fresh, RAPA-free medium for the next 24 h. In another set of experiments, cells from 12 de novo AML patients were treated with RAPA and Ara-C in above concentrations and time settings. RAPA and Ara-C were administrated to isolated peripheral blood mononuclear cells (PBMC). PBMC were immunophenotyped before and after treatment. Leukemic blasts were marked for individually chosen antigen, most characteristic for leukemic clone in particular patient. Normal CD3+ lymphocytes were also detected. Finally, Annexin V staining was performed. Based on that simultaneous three-color staining the proapoptotic effects of treatment could be measured by flow cytometry in both leukemic blasts and normal CD3+ cells. Thus, we found that pretreatment with RAPA protected majority of CD3+ cells (median of alive cells 85.5%) from Ara-C-induced apoptosis, whereas the leukemic blasts AI was higher than in samples treated with Ara-C. After Ara-C alone CD3+ rate decreased significantly (median 35.1%). Conclusions: Pretreatment with RAPA enhances cytotoxic effect of Ara-C on leukemic cells, but not on healthy lymphocytes. The phenomenon is probably due to reversible arrest of healthy cells in G1 phase of cell cycle by low doses of RAPA, what causes their transient resistance to proapoptotic action of cytostatic drugs. In contrast, the same RAPA doses selectively sensitizes leukemic cells to cytostatics. This suggests, that inhibition of mTOR kinase prior to cytostatics administration may result in selective anti-tumor treatment, with protection of normal cells.


2013 ◽  
Vol 41 (03) ◽  
pp. 615-628 ◽  
Author(s):  
Zengtao Xu ◽  
Guosheng Wu ◽  
Xu Wei ◽  
Xiuping Chen ◽  
Yitao Wang ◽  
...  

Celastrol is one of the principal active ingredients of Tripterygium wilfordii Hook.f., a toxic Chinese medical herb traditionally prescribed for controlling pain and inhibiting inflammation in various chronic inflammatory diseases, including rheumatoid arthritis (RA). Resistance to apoptosis of fibroblast-like synoviocytes is considered a major characteristic of RA. In this study, we test celastrol's cytotoxic effect and potential mechanisms in human rheumatoid synovial fibroblasts (RA-FLS). In the cytotoxic assay, we found that celastrol dose-dependently decreased RA-FLS viability and increased LDH release. The apoptotic nuclear morphology was observed after celastrol treatment as determined by DAPI fluorescence staining. Flow cytometry analysis with PI and Annexin V revealed that celastrol induced RA-FLS cell cycle arrest in the G2/M phase and apoptosis. Furthermore, celastrol dramatically increased expression of Bax/Bcl-2, proteolytic cleavage of Caspase-3, -9, PARP, and decreased expression of FasR. In addition, celastrol treatment resulted in DNA damage. Collectively, we concluded that celastrol inhibits RA-FLS proliferation by inducing DNA damage, cell cycle arrest, and apoptosis in vitro, which might provide data for its application in RA treatment.


2016 ◽  
Vol 64 (4) ◽  
pp. 957.1-957
Author(s):  
GC Osuji ◽  
M Reyes ◽  
NN Drever ◽  
TJ Kuehl ◽  
MN Uddin ◽  
...  

ObjectivePreeclampsia (preE) is a hypertensive disorder of pregnancy. Cardiotonic steroids (CTS) are endogenous inhibitors of Na+/K+ ATPase and at least one CTS, marinobufagenin (MBG), is elevated in preE prior to the development of the syndrome in rats with preE. MBG and ouabain impair cytotrophoblast (CTB) function, which is critical for placental development.Study DesignWe evaluated the effect of a CTS, cinobufotalin (CINO), on CTB cell function in vitro.ResultsCINO at ≥1 nM inhibited CTB cell proliferation, migration, and invasion (p<0.05) but had no effect on cell viability. There was a higher (p<0.05) percentage of G0/G1 phase cells in groups treated with CINO at ≥1 nM. CINO caused an increase in stress signaling p38 MAPK and a positive annexin-V staining in CTB cells, indicating the activation of apoptotic signaling. However, the CINO induced apoptotic signaling was prevented by p38 inhibition.ConclusionThis data demonstrates that CINO impairs CTB cell function via cell cycle arrest and apoptotic signaling.


2021 ◽  
Author(s):  
Heba Sahyon ◽  
Eman N.M. Ramadan ◽  
Fayez Althobaiti ◽  
Mohammad M.A. Mashaly

Abstract Combination therapy comprising natural polyphenols and anticancer drugs has been used to decrease the adverse effects and increase the effectiveness and antioxidant activities of the drugs. The antioxidant and anticancer effects of quercetin (Q), a nutritive polyphenol, have been observed both in vitro and in vivo. Likewise, the anticancer activity of sulfamethoxazole (S) has been demonstrated in vitro and in vivo. This study aimed to investigate the in vitro and in vivo anticancer effects of Q alone and in combination with S. The in vitro effects of S, Q, and S + Q on HCT-116, HepG2, MCF-7, and PC3 cell lines were examined. Additionally, the in vivo effects of these drugs were evaluated using Ehrlich ascites carcinoma (EAC) tumor-bearing mice. The in vitro data revealed the potent anticancer activity of S + Q through the induction of apoptosis and cell cycle arrest. The EAC-inoculated mice treated with S + Q presented with elevated SOD, GSH, CAT, and TAC levels and decreased malondialdehyde levels compared with the untreated EAC group, thus revealing the antioxidant and protective actions of S + Q against EAC cells invasion. Furthermore, the downregulation of NFkB and upregulation of the caspase3 gene in the EAC-inoculated mice treated with the S + Q indicated the induction of the apoptotic pathway and decrease in both cell proliferation and metastasis. In conclusion, the combination of S and Q might exert anticancer effects by inducing apoptosis and exhibiting selective toxicity against the cancer cells and thereby protecting the vital organs.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Xiangnan Zhao ◽  
Yue Liu ◽  
Pingping Jia ◽  
Hui Cheng ◽  
Chen Wang ◽  
...  

Abstract Background The senescence of dermal fibroblasts (DFLs) leads to an imbalance in the synthesis and degradation of extracellular matrix (ECM) proteins, presenting so-called senescence-associated secretory phenotype (SASP), which ultimately leads to skin aging. Recently, mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have been recognized as a promising cell-free therapy for degenerative diseases, which opens a new avenue for skin aging treatment. Methods In this study, we utilized chitosan (CS) hydrogel for effective loading and sustained release of EVs. In vitro, we explored the rejuvenation effects of CS hydrogel-incorporated EVs (CS-EVs) on replicative senescence DFLs through a series of experiments such as senescence-associated β-galactosidase (SA-β-gal) staining, RT-PCR, and Western blot analysis. Besides, we employed local multi-site subcutaneous injection to treat skin aging of naturally aged mice with CS-EVs and DiI fluorescent dye was used to label EVs to achieve in vivo real-time tracking. Results CS-EVs can significantly improve the biological functions of senescent fibroblasts, including promoting their proliferation, enhancing the synthesis of ECM proteins, and inhibiting the overexpression of matrix metalloproteinases (MMPs). Moreover, CS hydrogel could prolong the release of EVs and significantly increase the retention of EVs in vivo. After CS-EVs subcutaneous injection treatment, the aging skin tissues showed a rejuvenation state, manifested explicitly as the enhanced expression of collagen, the decreased expression of SASP-related factors, and the restoration of tissue structures. Conclusions CS hydrogel-encapsulated EVs could delay the skin aging processes by ameliorating the function of aging DFLs. Our results also highlight the potential of CS hydrogel-encapsulated EVs as a novel therapeutic strategy for improving aging skin to rejuvenation.


Cells ◽  
2019 ◽  
Vol 8 (12) ◽  
pp. 1546 ◽  
Author(s):  
Diane Wengerodt ◽  
Christian Schmeer ◽  
Otto W. Witte ◽  
Alexandra Kretz

Replicative senescence has initially been defined as a stress reaction of replication-competent cultured cells in vitro, resulting in an ultimate cell cycle arrest at preserved growth and viability. Classically, it has been linked to critical telomere curtailment following repetitive cell divisions, and later described as a response to oncogenes and other stressors. Currently, there are compelling new directions indicating that a comparable state of cellular senescence might be adopted also by postmitotic cell entities, including terminally differentiated neurons. However, the cellular upstream inducers and molecular downstream cues mediating a senescence-like state in neurons (amitosenescence) are ill-defined. Here, we address the phenomenon of abortive atypical cell cycle activity in light of amitosenescence, and discuss why such replicative reprogramming might provide a yet unconsidered source to explain senescence in maturated neurons. We also hypothesize the existence of a G0 subphase as a priming factor for cell cycle re-entry, in analogy to discoveries in quiescent muscle stem cells. In conclusion, we propose a revision of our current view on the process and definition of senescence by encompassing a primarily replication-incompetent state (amitosenescence), which might be expanded by events of atypical cell cycle activity (pseudomitosenescence).


2019 ◽  
Vol 20 (5) ◽  
pp. 1217 ◽  
Author(s):  
Bresler Swanepoel ◽  
Luanne Venables ◽  
Octavian Olaru ◽  
George Nitulescu ◽  
Maryna van de Venter

Anemone nemorosa is part of the Ranunculaceae genus Anemone (order Ranunculales) which comprises more than 150 species. Various parts of the plant have been used for the treatment of numerous medical conditions such as headaches, tertian agues, rheumatic gout, leprosy, lethargy, eye inflammation as well as malignant and corroding ulcers. The Anemone plants have been found to contain various medicinal compounds with anti-cancer, immunomodulatory, anti-inflammatory, anti-oxidant and anti-microbial activities. To date there has been no reported evidence of its use in the treatment of cancer. However, due to the reported abundance of saponins which usually exert anti-cancer activity via cell cycle arrest and the induction of apoptosis, we investigated the mode of cell death induced by an aqueous A. nemorosa extract by using HeLa cervical cancer cells. Cisplatin was used as a positive control. With a 50% inhibitory concentration (IC50) of 20.33 ± 2.480 µg/mL, treatment with A. nemorosa yielded a delay in the early mitosis phase of the cell cycle. Apoptosis was confirmed through fluorescent staining with annexin V-FITC. Apoptosis was more evident with A. nemorosa treatment compared to the positive control after 24 and 48 h. Tetramethylrhodamine ethyl ester staining showed a decrease in mitochondrial membrane potential at 24 and 48 h. The results obtained imply that A. nemorosa may have potential anti-proliferative properties.


Sign in / Sign up

Export Citation Format

Share Document