The multifunctional BAG-1 (Bcl-2 athanogene-1) protein promotes breast cancer survival through direct or indirect interaction partners. The number of the interacting partners determines its cellular role in different conditions. As well as interaction partner variability, the amount of BAG-1 protein in the cells could cause dramatic alterations. According to previous studies, while the transient silencing of Bag-1 enhanced drug-induced apoptosis, deletion of BAG-1 could induce stemness properties and Akt-mediated actin remodeling in MCF-7 breast cancer cells. Considering the heterogeneity of breast cancer and the variability of BAG-1 -mediated cell response, it has become essential to determine microRNA (miRNA) functions in breast cancer depending on Bag-1 expression level. This study aims to compare microRNA expression levels in wt and Bag-1 knockout (KO) MCF-7 breast cancer cells. hsa-miR-429 was selected as a potential miRNA in BAG-1KO MCF-7 cells because of the downregulation both in bioinformatics and validation qRT-PCR assay. According to predicted mRNA targets and functional enrichment analysis the ten hub proteins that are phosphatidylinositol-4,5-biphosphate 3-kinase catalytic subunit alpha (PIK3CA), kinase insert domain receptor (KDR), GRB2 associated binding protein 1 (GAB1), Rac family small GTPase1 (RAC1), vascular endothelial growth factor A (VEGFA), Cbl proto-oncogene (CBL), syndecan 2 (SDC2), phospholipase C gamma 1 (PLCG1), E1A binding protein p300 (EP300), and CRK like proto-oncogene, adaptor protein (CRKL) were identified as targets of hsa-miR-429. The functional enrichment analysis showed that the most significant proteins were enriched in PI3K/Akt, focal adhesion, cytoskeleton regulation, proteoglycans in cancer, and Ras signaling pathways. It was determined that hsa-miR-429 targeted these pathways in Bag-1 deficient conditions and could be used as a potential therapeutic target in future studies.
{"title":"The comparison of differentially expressed microRNAs in Bag-1 deficient and wild type MCF-7 breast cancer cells by small RNA sequencing.","authors":"Pelin Özfiliz Kilbaş, Gizem Alkurt, Pinar Obakan Yerlikaya, Ajda Çoker Gürkan, Gizem Dinler Doğanay, Elif Damla Arisan","doi":"10.3906/biy-2109-48","DOIUrl":"https://doi.org/10.3906/biy-2109-48","url":null,"abstract":"<p><p>The multifunctional BAG-1 (Bcl-2 athanogene-1) protein promotes breast cancer survival through direct or indirect interaction partners. The number of the interacting partners determines its cellular role in different conditions. As well as interaction partner variability, the amount of BAG-1 protein in the cells could cause dramatic alterations. According to previous studies, while the transient silencing of Bag-1 enhanced drug-induced apoptosis, deletion of BAG-1 could induce stemness properties and Akt-mediated actin remodeling in MCF-7 breast cancer cells. Considering the heterogeneity of breast cancer and the variability of BAG-1 -mediated cell response, it has become essential to determine microRNA (miRNA) functions in breast cancer depending on <i>Bag-1</i> expression level. This study aims to compare microRNA expression levels in wt and <i>Bag-1</i> knockout (KO) MCF-7 breast cancer cells. hsa-miR-429 was selected as a potential miRNA in BAG-1KO MCF-7 cells because of the downregulation both in bioinformatics and validation qRT-PCR assay. According to predicted mRNA targets and functional enrichment analysis the ten hub proteins that are phosphatidylinositol-4,5-biphosphate 3-kinase catalytic subunit alpha (PIK3CA), kinase insert domain receptor (KDR), GRB2 associated binding protein 1 (GAB1), Rac family small GTPase1 (RAC1), vascular endothelial growth factor A (VEGFA), Cbl proto-oncogene (CBL), syndecan 2 (SDC2), phospholipase C gamma 1 (PLCG1), E1A binding protein p300 (EP300), and CRK like proto-oncogene, adaptor protein (CRKL) were identified as targets of hsa-miR-429. The functional enrichment analysis showed that the most significant proteins were enriched in PI3K/Akt, focal adhesion, cytoskeleton regulation, proteoglycans in cancer, and Ras signaling pathways. It was determined that hsa-miR-429 targeted these pathways in Bag-1 deficient conditions and could be used as a potential therapeutic target in future studies.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 2","pages":"118-136"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10393109/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10307890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background/aim: Icariin (ICA)-loaded zein/PLGA nanofiber membrane combined with MSCs was prepared by coaxial electrospinning and bioelectrospraying.
Materials and methods: SEM and TEM were used to evaluate the surface morphology and microstructure of the fiber membrane. Ultraviolet spectrophotometry was used to detect drug release. A LIVE/DEAD Viability/Cytotoxicity Kit and fluorescence staining were used to detect cell morphology and activity. Alkaline phosphatase and calcium mineralization deposition were used to evaluate the osteoinductive activity of the scaffold. Dynamic mechanical analysis was used to determine the Young's modulus, maximum load, and maximum elongation of the prepared scaffold. Western blot was used to detect the related protein expression in MSCs induced by drug-loaded scaffolds.
Results: Good mechanical properties and stability were observed in the prepared drug-loaded scaffolds. SEM showed that there were a considerable number of MSCs dispersed in the scaffold. MSCs were evenly distributed, could grow evenly between fibers, and were arranged orderly along the fibers. Sustained release of ICA confers cell laden scaffold higher ALP activity and ECM mineral deposition through Runx2, OPN and OCN pathways.
Conclusion: Isotropic sustained release of ICA grant cell laden scaffolds rapid bone regeneration compacity which can provide a good osteogenic environment for loaded MSCs.
{"title":"Three-dimensional mesenchymal stem cell laden scaffold of icariin sustained-release for bone regeneration.","authors":"Yanbing Liu, Yan Fang","doi":"10.55730/1300-0152.2627","DOIUrl":"https://doi.org/10.55730/1300-0152.2627","url":null,"abstract":"<p><strong>Background/aim: </strong>Icariin (ICA)-loaded zein/PLGA nanofiber membrane combined with MSCs was prepared by coaxial electrospinning and bioelectrospraying.</p><p><strong>Materials and methods: </strong>SEM and TEM were used to evaluate the surface morphology and microstructure of the fiber membrane. Ultraviolet spectrophotometry was used to detect drug release. A LIVE/DEAD Viability/Cytotoxicity Kit and fluorescence staining were used to detect cell morphology and activity. Alkaline phosphatase and calcium mineralization deposition were used to evaluate the osteoinductive activity of the scaffold. Dynamic mechanical analysis was used to determine the Young's modulus, maximum load, and maximum elongation of the prepared scaffold. Western blot was used to detect the related protein expression in MSCs induced by drug-loaded scaffolds.</p><p><strong>Results: </strong>Good mechanical properties and stability were observed in the prepared drug-loaded scaffolds. SEM showed that there were a considerable number of MSCs dispersed in the scaffold. MSCs were evenly distributed, could grow evenly between fibers, and were arranged orderly along the fibers. Sustained release of ICA confers cell laden scaffold higher ALP activity and ECM mineral deposition through Runx2, OPN and OCN pathways.</p><p><strong>Conclusion: </strong>Isotropic sustained release of ICA grant cell laden scaffolds rapid bone regeneration compacity which can provide a good osteogenic environment for loaded MSCs.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 5","pages":"414-425"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388105/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9922858","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Accumulating evidence shows that gut microbial dysbiosis may represent a risk factor for Parkinson's disease (PD). Exercise has a positive effect on microbiota in general. The effect of aerobic exercise training (AET) on the gut microbial environment in PD remains to be explored. Here, we performed the 16S rRNA gene sequencing on feces from sham operated-mice (sham), PD mice model, and mice receiving AET (AET). Results indicated that AET had no remarkable effect on species richness and bacterial diversity of PD mice. The relative abundance of the Bacteroidetes was reduced, while Firmicutes, Actinobacteria, Lactobacillaceae, Streptococcaceae, Lactobacillus, Streptococcus, Lactococcus, Lysinibacillus, Pelomonas, and Prevotellaceae_UCG-001 were increased in PD mice compared with those of sham operated-mice, whereas AET partly rescued their abundance. Additionally, the composition proportion of beneficial Lactobacillus_gasseri and uncultured_Erysipelotrichales_bacterium significantly increased in AET mice compared to PD mice. Moreover, discriminative bacteria, such as Bacilli, Lactobacillales, Lactobacillaceae, Lactobacillus, and Lactococcus were identified as a specific taxon in AET mice. Here we provide evidence that AET can improve the gut microbiota of PD mice.
{"title":"Influence of aerobic exercise training on mice gut microbiota in Parkinson's disease.","authors":"Tianlun Fan, Xiating Li, Xiang Zhang, Jing Zhang, Lichun Sun, Jingjing Chen, Chuan Fu","doi":"10.55730/1300-0152.2617","DOIUrl":"https://doi.org/10.55730/1300-0152.2617","url":null,"abstract":"<p><p>Accumulating evidence shows that gut microbial dysbiosis may represent a risk factor for Parkinson's disease (PD). Exercise has a positive effect on microbiota in general. The effect of aerobic exercise training (AET) on the gut microbial environment in PD remains to be explored. Here, we performed the 16S rRNA gene sequencing on feces from sham operated-mice (sham), PD mice model, and mice receiving AET (AET). Results indicated that AET had no remarkable effect on species richness and bacterial diversity of PD mice. The relative abundance of the <i>Bacteroidetes</i> was reduced, while <i>Firmicutes</i>, <i>Actinobacteria</i>, <i>Lactobacillaceae</i>, <i>Streptococcaceae</i>, <i>Lactobacillus</i>, <i>Streptococcus</i>, <i>Lactococcus</i>, <i>Lysinibacillus</i>, <i>Pelomonas</i>, and <i>Prevotellaceae_UCG-001</i> were increased in PD mice compared with those of sham operated-mice, whereas AET partly rescued their abundance. Additionally, the composition proportion of beneficial <i>Lactobacillus_gasseri</i> and u<i>ncultured_Erysipelotrichales_bacterium</i> significantly increased in AET mice compared to PD mice. Moreover, discriminative bacteria, such as <i>Bacilli</i>, <i>Lactobacillales</i>, <i>Lactobacillaceae</i>, <i>Lactobacillus</i>, and <i>Lactococcus</i> were identified as a specific taxon in AET mice. Here we provide evidence that AET can improve the gut microbiota of PD mice.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 4","pages":"288-297"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388120/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9925975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Particular somatic cells, namely cumulus cells (CCs) that support the oocyte maturation, fertility, and viability by providing the nutrients and energy to the oocyte envelop the mammalian oocyte. In this study, discarded human cumulus tissues were used to reveal the value of hyaluronic acid-rich CCs on several cellular events, including differentiation. Conditioned media, recovered from the primary culture of CCs, were introduced to the human nucleus pulposus cells (hNPCs) which were functionally distorted because of the loss of chondrogenecity. Enlightening the impact of cumulus conditioned media (CCM) on wound healing and angiogenesis was also investigated. In line with these goals, differentiation of hNPCs into chondrocytes with CCM as the basal medium containing traditional differentiation agents was induced upon isolation and characterization of hCCs and hNPCs. The effects were detected by differentiation-specific cell stains and gene expression analyses. Scratch and tube formation assays were performed to detect the effect of CCM on wound healing and angiogenesis. Our results showed that cumulus cell-conditioned media promoted the chondrogenesis and osteogenesis of hNPCs. A significant increase in angiogenesis and ability for wound closure was detected only in groups cultured in CCM compared to the control. These findings demonstrated that CCM might be used in therapeutics.
{"title":"The investigation of diverse physiological and therapeutic impact of cellular-based products derived from human cumulus cells.","authors":"Derya Burukçu, Sema Yilmaz, Oya Alagöz, Cumhur Kaan Yaltirik, Erkut Attar, Fikrettin Şahin, Esra Aydemir","doi":"10.55730/1300-0152.2626","DOIUrl":"https://doi.org/10.55730/1300-0152.2626","url":null,"abstract":"<p><p>Particular somatic cells, namely cumulus cells (CCs) that support the oocyte maturation, fertility, and viability by providing the nutrients and energy to the oocyte envelop the mammalian oocyte. In this study, discarded human cumulus tissues were used to reveal the value of hyaluronic acid-rich CCs on several cellular events, including differentiation. Conditioned media, recovered from the primary culture of CCs, were introduced to the human nucleus pulposus cells (hNPCs) which were functionally distorted because of the loss of chondrogenecity. Enlightening the impact of cumulus conditioned media (CCM) on wound healing and angiogenesis was also investigated. In line with these goals, differentiation of hNPCs into chondrocytes with CCM as the basal medium containing traditional differentiation agents was induced upon isolation and characterization of hCCs and hNPCs. The effects were detected by differentiation-specific cell stains and gene expression analyses. Scratch and tube formation assays were performed to detect the effect of CCM on wound healing and angiogenesis. Our results showed that cumulus cell-conditioned media promoted the chondrogenesis and osteogenesis of hNPCs. A significant increase in angiogenesis and ability for wound closure was detected only in groups cultured in CCM compared to the control. These findings demonstrated that CCM might be used in therapeutics.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 5","pages":"400-413"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388000/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9926249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fat mass and obesity-associated protein (FTO) is a demethylase and plays a vital role in various cancers. However, the regulation mechanism of FTO in prostate cancer (PCa) remains unclear. This study aimed to elucidate the mechanism of FTO in PCa. The function and mechanism of FTO-mediated in PCa were determined by gain-of-function assays and RNA-seq. We found that FTO expression in PCa tissues and two PCa cell lines were significantly lower than that in adjacent tissues and normal cell line. PCa cells after overexpression of FTO showed a significant lower in proliferation, migration, and invasion capabilities. RNA-seq displayed that FTO overexpression altered transcriptome landscape in Du145 and PC-3 cells, particularly upregulating EGR2 expression. FTO overexpression induced differential expression genes, including MYLK2, DNA2, CDK, and CDC (6, 7, 20, 25, and 45), which were mainly enriched in adjustment of cell cycle and growth pathways. Furthermore, FTO overexpression significantly reduced the EGR2 methylation level. Arresting the proliferation, migration, and invasion of Du145 cells induced by FTO overexpression was significantly rescued by EGR2 knockdown. FTO overexpression also significantly inhibited tumor growth and promoted EGR2 protein expression. Taken together, FTO suppresses PCa progression by regulating EGR2 methylation. We uncovered a novel regulatory mechanism of FTO in PCa and provide a new potential therapeutic target for PCa.
{"title":"Demethylase FTO inhibits the development of prostate cancer by upregulating EGR2 expression in an m6A manner.","authors":"Zhenyu Wang, Huamin Sun, Hua Zhu, Donghua Gu, Xinfeng Chen, Yongsheng Pan, Bing Zheng, Dongrong Yang","doi":"10.55730/1300-0152.2629","DOIUrl":"https://doi.org/10.55730/1300-0152.2629","url":null,"abstract":"<p><p>Fat mass and obesity-associated protein (FTO) is a demethylase and plays a vital role in various cancers. However, the regulation mechanism of FTO in prostate cancer (PCa) remains unclear. This study aimed to elucidate the mechanism of FTO in PCa. The function and mechanism of FTO-mediated in PCa were determined by gain-of-function assays and RNA-seq. We found that FTO expression in PCa tissues and two PCa cell lines were significantly lower than that in adjacent tissues and normal cell line. PCa cells after overexpression of FTO showed a significant lower in proliferation, migration, and invasion capabilities. RNA-seq displayed that FTO overexpression altered transcriptome landscape in Du145 and PC-3 cells, particularly upregulating EGR2 expression. FTO overexpression induced differential expression genes, including MYLK2, DNA2, CDK, and CDC (6, 7, 20, 25, and 45), which were mainly enriched in adjustment of cell cycle and growth pathways. Furthermore, FTO overexpression significantly reduced the EGR2 methylation level. Arresting the proliferation, migration, and invasion of Du145 cells induced by FTO overexpression was significantly rescued by EGR2 knockdown. FTO overexpression also significantly inhibited tumor growth and promoted EGR2 protein expression. Taken together, FTO suppresses PCa progression by regulating EGR2 methylation. We uncovered a novel regulatory mechanism of FTO in PCa and provide a new potential therapeutic target for PCa.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 6","pages":"426-438"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10387994/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9926778","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The prognostic characteristics of pancreatic cancer (PC) are determined by the contributing factors from the tumor microenvironment. Leptin is a critical oncogenic factor released by adipocytes as an adipokine into the tumor microenvironment, where it promotes tumor development by activating cancer stem cell (CSC) molecular regulators Notch, Hedgehog, and Wnt/β-catenin signaling. One of the downstream targets of these pathways is CDK4/6 and cyclin D which is controlled by P16 INK4A that is highly mutated in PC. Therefore, the purpose of this study was to determine the effect of a CDK4/6 inhibitor, palbociclib, on Leptin-induced PC cells and to target the Notch, Hedgehog, and Wnt/β-catenin signaling pathways via miR-150, miR-506, and miR-208 modulation. Leptin treatment increased the ability of Panc-1, MiaPaCa-2, and Capan-2 cells to proliferate and decreased the effect of palbociclib. Additionally, tumorspheres were generated from Leptin-treated (Leptin+) and Leptin-untreated (Leptin-) Panc-1 and MiaPaCa-2 cells and transfected with miR-506, miR-150 (tumorsuppressor miRNAs), or anti-miR-208 (oncomiR), followed by palbociclib treatment. Forced expression of miR-506 or miR-150 significantly increased the susceptibility of Leptin+ cells to palbociclib treatment by inhibiting colony and tumor spheroid formation, and CD44 expression in Panc-1 and MiaPaCa-2 cells. Additionally, the increased miR-150 expression is more effective at inhibiting N-cadherin, β-catenin, p-GSK3β, Notch, and Wnt5a/b expression in Leptin-/+ Panc-1 and MiaPaCa-2 cells. As a result, palbociclib suppressed the CSC profile induced by leptin treatment, inhibiting both tumorsphere forms and leptin-targeted signaling pathways, thereby disabling the Panc-1 and MiaPaCa-2 cells' resistance mechanism. Increased expression of miR-506 or miR-150 and inhibition of miR-208 enhanced sensitivity of Panc-1 and MiaPaCa-2 Leptin-/+ cells to palbociclib treatment. As a result, this study proved that combining inhibitors of CSC molecular regulators with palbociclib improves the success rate of inhibition of PC cell proliferation.
{"title":"Palbociclib suppresses the cancer stem cell properties and cell proliferation through increased levels of miR-506 or miR-150 in Panc-1 and MiaPaCa-2 cells.","authors":"Özge Rencüzoğullari, Elif Damla Arisan","doi":"10.55730/1300-0152.2622","DOIUrl":"https://doi.org/10.55730/1300-0152.2622","url":null,"abstract":"<p><p>The prognostic characteristics of pancreatic cancer (PC) are determined by the contributing factors from the tumor microenvironment. Leptin is a critical oncogenic factor released by adipocytes as an adipokine into the tumor microenvironment, where it promotes tumor development by activating cancer stem cell (CSC) molecular regulators Notch, Hedgehog, and Wnt/β-catenin signaling. One of the downstream targets of these pathways is CDK4/6 and cyclin D which is controlled by P16 INK4A that is highly mutated in PC. Therefore, the purpose of this study was to determine the effect of a CDK4/6 inhibitor, palbociclib, on Leptin-induced PC cells and to target the Notch, Hedgehog, and Wnt/β-catenin signaling pathways via miR-150, miR-506, and miR-208 modulation. Leptin treatment increased the ability of Panc-1, MiaPaCa-2, and Capan-2 cells to proliferate and decreased the effect of palbociclib. Additionally, tumorspheres were generated from Leptin-treated (Leptin+) and Leptin-untreated (Leptin-) Panc-1 and MiaPaCa-2 cells and transfected with miR-506, miR-150 (tumorsuppressor miRNAs), or anti-miR-208 (oncomiR), followed by palbociclib treatment. Forced expression of miR-506 or miR-150 significantly increased the susceptibility of Leptin+ cells to palbociclib treatment by inhibiting colony and tumor spheroid formation, and CD44 expression in Panc-1 and MiaPaCa-2 cells. Additionally, the increased miR-150 expression is more effective at inhibiting N-cadherin, β-catenin, p-GSK3β, Notch, and Wnt5a/b expression in Leptin-/+ Panc-1 and MiaPaCa-2 cells. As a result, palbociclib suppressed the CSC profile induced by leptin treatment, inhibiting both tumorsphere forms and leptin-targeted signaling pathways, thereby disabling the Panc-1 and MiaPaCa-2 cells' resistance mechanism. Increased expression of miR-506 or miR-150 and inhibition of miR-208 enhanced sensitivity of Panc-1 and MiaPaCa-2 Leptin-/+ cells to palbociclib treatment. As a result, this study proved that combining inhibitors of CSC molecular regulators with palbociclib improves the success rate of inhibition of PC cell proliferation.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 5","pages":"342-360"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10387936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9928293","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pınar Obakan Yerlikaya, Leila Mehdizadehtapeh, Özge Rencüzoğullari, Fadina Kuryayeva, Sena Sedef Çevikli, Şevval Özağar, Sibel Pınar Odabaş, Sude Tunçkol, Hakan Yetim, Ajda Çoker Gürkan, Elif Damla Arisan
Gemcitabine is a broad-spectrum antimetabolite and a deoxycytidine analog recognized as a standard therapy alone or in combination with other antineoplastic agents in the therapy of pancreas cancer. Drug resistance following gemcitabine treatment is a common phenomenon; therefore, combinational therapy models are usually preferred. Pancreatic ductal adenocarcinoma, or pancreas cancer, is the fourth leading cause of cancer-related deaths worldwide. With the increasing incidence of pancreatic cancer every year, the mortality rate is also rising significantly because of late diagnosis, and limited chemotherapy options. Adjuvant chemotherapy after surgical resection is the typical option for the treatment of early pancreatic cancer. Mostly, 5-fluorouracil/leucovorin with irinotecan and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel is used for the prognosis of advanced pancreatic cancer; however, chemoresistance usually occurs limiting the effectiveness of the chemotherapy. Therefore, most of the studies are focused on gemcitabine combination with other drugs to overcome the situation. As an apoptotic agent and a member of brassinosteroids, epibrassinolide (EBR) induces endoplasmic reticulum (ER) stress-dependent cell death in different cancer cells, as shown by our group. In this study, we aimed to enhance the gemcitabine apoptotic effect by EBR combined treatment in pancreatic cancer cells. EBR treatment reduced cell viability and inhibited cell proliferation in PANC-1, MIA PaCa-2, and AsPC-1 cells. Each pancreatic cancer cell gave different responses to the EBR treatment because of different aggressiveness. However, EBR induced apoptosis through increasing ROS generation, which was associated with ER stress in PANC-1 and MIA PaCa-2 cells. Gemcitabine alone reduced the cell viability of each pancreatic cancer cell line; however, combination with EBR led to further induction of apoptotic cell death in each pancreatic cancer cell line. In addition, combined treatment of gemcitabine and EBR further decreased N-cadherin and vimentin expressions, suggesting that epithelial-mesenchymal transition of pancreatic cells is reduced. In conclusion, EBR had therapeutic potential to avoid the gemcitabine-induced side effects during the treatment of pancreatic cancer.
{"title":"Gemcitabine in combination with epibrassinolide enhanced the apoptotic response in an ER stress-dependent manner and reduced the epithelial-mesenchymal transition in pancreatic cancer cells.","authors":"Pınar Obakan Yerlikaya, Leila Mehdizadehtapeh, Özge Rencüzoğullari, Fadina Kuryayeva, Sena Sedef Çevikli, Şevval Özağar, Sibel Pınar Odabaş, Sude Tunçkol, Hakan Yetim, Ajda Çoker Gürkan, Elif Damla Arisan","doi":"10.55730/1300-0152.2630","DOIUrl":"https://doi.org/10.55730/1300-0152.2630","url":null,"abstract":"<p><p>Gemcitabine is a broad-spectrum antimetabolite and a deoxycytidine analog recognized as a standard therapy alone or in combination with other antineoplastic agents in the therapy of pancreas cancer. Drug resistance following gemcitabine treatment is a common phenomenon; therefore, combinational therapy models are usually preferred. Pancreatic ductal adenocarcinoma, or pancreas cancer, is the fourth leading cause of cancer-related deaths worldwide. With the increasing incidence of pancreatic cancer every year, the mortality rate is also rising significantly because of late diagnosis, and limited chemotherapy options. Adjuvant chemotherapy after surgical resection is the typical option for the treatment of early pancreatic cancer. Mostly, 5-fluorouracil/leucovorin with irinotecan and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel is used for the prognosis of advanced pancreatic cancer; however, chemoresistance usually occurs limiting the effectiveness of the chemotherapy. Therefore, most of the studies are focused on gemcitabine combination with other drugs to overcome the situation. As an apoptotic agent and a member of brassinosteroids, epibrassinolide (EBR) induces endoplasmic reticulum (ER) stress-dependent cell death in different cancer cells, as shown by our group. In this study, we aimed to enhance the gemcitabine apoptotic effect by EBR combined treatment in pancreatic cancer cells. EBR treatment reduced cell viability and inhibited cell proliferation in PANC-1, MIA PaCa-2, and AsPC-1 cells. Each pancreatic cancer cell gave different responses to the EBR treatment because of different aggressiveness. However, EBR induced apoptosis through increasing ROS generation, which was associated with ER stress in PANC-1 and MIA PaCa-2 cells. Gemcitabine alone reduced the cell viability of each pancreatic cancer cell line; however, combination with EBR led to further induction of apoptotic cell death in each pancreatic cancer cell line. In addition, combined treatment of gemcitabine and EBR further decreased N-cadherin and vimentin expressions, suggesting that epithelial-mesenchymal transition of pancreatic cells is reduced. In conclusion, EBR had therapeutic potential to avoid the gemcitabine-induced side effects during the treatment of pancreatic cancer.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 6","pages":"439-457"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10387961/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9929303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Salwa A Fouad, Gamal H El-Sokkary, Abo Bakr Abdel Shakor
Arabinofuranosyl cytidine (AraC) is an analog of deoxycytidine used as an anticancer drug for leukemic patients. The effective dose always produces severe complications. The present study investigated the modulation of autophagy and its impact on the cytotoxicity of AraC toward murine myelogenous leukemia cells (M-NFS-60). Autophagy was inhibited by NH4Cl or Bafilomycin A1 or enhanced by amino acid starvation, glucose starvation, mild hyperthermia (41 °C), or rapamycin (Rap). Cells were treated with different concentrations, 0 to 2 μM, of AraC in the presence or absence of autophagy modulators. AraC-induced apoptosis is combined with autophagy, especially at lower concentrations. This autophagy is characterized by a slow flux, as indicated by levels of LC3B II and P62 proteins. Inhibition of autophagy did not alter cleaved caspase 3 levels (c-casp.3) or cell viability measured by MTT assays. Conversely, acceleration of AraC-induced autophagy by co-treatment with autophagy inducers reduced cell viability and increased c-casp.3 and c-PARP levels. Further, c-PARP levels were reduced in the presence of caspase inhibitor, Z-VAD-FMK. Enhancement of slow autophagic flux induced by low concentrations of AraC significantly increased the cytotoxicity of AraC toward M-NFS-60 cells. Such coadministration of autophagy inducers might improve the efficacy of AraC treatment and reduce effective doses.
{"title":"Acceleration of slow autophagy flux induced by arabinofuranosyl cytidine improves its antileukemic effectiveness in M-NFS-60 cells.","authors":"Salwa A Fouad, Gamal H El-Sokkary, Abo Bakr Abdel Shakor","doi":"10.55730/1300-0152.2619","DOIUrl":"https://doi.org/10.55730/1300-0152.2619","url":null,"abstract":"<p><p>Arabinofuranosyl cytidine (AraC) is an analog of deoxycytidine used as an anticancer drug for leukemic patients. The effective dose always produces severe complications. The present study investigated the modulation of autophagy and its impact on the cytotoxicity of AraC toward murine myelogenous leukemia cells (M-NFS-60). Autophagy was inhibited by NH<sub>4</sub>Cl or Bafilomycin A1 or enhanced by amino acid starvation, glucose starvation, mild hyperthermia (41 °C), or rapamycin (Rap). Cells were treated with different concentrations, 0 to 2 μM, of AraC in the presence or absence of autophagy modulators. AraC-induced apoptosis is combined with autophagy, especially at lower concentrations. This autophagy is characterized by a slow flux, as indicated by levels of LC3B II and P62 proteins. Inhibition of autophagy did not alter cleaved caspase 3 levels (c-casp.3) or cell viability measured by MTT assays. Conversely, acceleration of AraC-induced autophagy by co-treatment with autophagy inducers reduced cell viability and increased c-casp.3 and c-PARP levels. Further, c-PARP levels were reduced in the presence of caspase inhibitor, Z-VAD-FMK. Enhancement of slow autophagic flux induced by low concentrations of AraC significantly increased the cytotoxicity of AraC toward M-NFS-60 cells. Such coadministration of autophagy inducers might improve the efficacy of AraC treatment and reduce effective doses.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 4","pages":"307-317"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388043/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9922869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Parkinson's disease (PD) is a common complex neurodegenerative disease, and aerobic exercise (EX) has potential to improve motor dysfunction. This study aimed to explore whether EX acts on PD in mice mode. Mice were administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and subjected to a 4-week physical exercise regimen (EX-PD group) and underwent RNA-Seq. Here, MPTP caused PD, which was characterized by neuron shrinkage and behavioral deficits, whereas EX improved PD by rescuing neuronal survival and motor function in mice. Moreover, circRNA expression profiles identified a total of 142 differentially expressed circRNAs (DEcircRNAs) between PD and EX-PD group. These DEcircRNAs were mainly involved in PD, dopaminergic synapses, and calcium signaling pathways. The expression of circZfp827 and circTshz2 were significantly elevated in PD group while reduced owing to EX intervention. In contrast, EX intervention significantly restored decline in circHivep2 expression due to PD. The circRNA-miRNA-mRNA network suggested that circZfp827, circHivep2, and circTshz2 were involved in ceRNA mechanism of EX to improve PD, and their target genes were significantly decreased after interference. The directly binding regulation relationship of circTshz2-mmu-miR-326-3p-Th was verified by double luciferase reporter assay. Our research revealed that EX improved motor behavioral deficits and pathological features of PD mice, and circRNA-based signatures are potential candidates for further assessment as PD biomarkers for improvement by EX.
{"title":"Transcriptome sequencing reveals circRNA expression profile in Parkinson's disease-like mice after aerobic exercise.","authors":"Tianlun Fan, Xiating Li, Chuan Fu, Lichun Sun","doi":"10.55730/1300-0152.2611","DOIUrl":"https://doi.org/10.55730/1300-0152.2611","url":null,"abstract":"<p><p>Parkinson's disease (PD) is a common complex neurodegenerative disease, and aerobic exercise (EX) has potential to improve motor dysfunction. This study aimed to explore whether EX acts on PD in mice mode. Mice were administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and subjected to a 4-week physical exercise regimen (EX-PD group) and underwent RNA-Seq. Here, MPTP caused PD, which was characterized by neuron shrinkage and behavioral deficits, whereas EX improved PD by rescuing neuronal survival and motor function in mice. Moreover, circRNA expression profiles identified a total of 142 differentially expressed circRNAs (DEcircRNAs) between PD and EX-PD group. These DEcircRNAs were mainly involved in PD, dopaminergic synapses, and calcium signaling pathways. The expression of circZfp827 and circTshz2 were significantly elevated in PD group while reduced owing to EX intervention. In contrast, EX intervention significantly restored decline in circHivep2 expression due to PD. The circRNA-miRNA-mRNA network suggested that circZfp827, circHivep2, and circTshz2 were involved in ceRNA mechanism of EX to improve PD, and their target genes were significantly decreased after interference. The directly binding regulation relationship of circTshz2-mmu-miR-326-3p-Th was verified by double luciferase reporter assay. Our research revealed that EX improved motor behavioral deficits and pathological features of PD mice, and circRNA-based signatures are potential candidates for further assessment as PD biomarkers for improvement by EX.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 3","pages":"227-238"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10388083/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9922886","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Özgün Fırat Düzenli, Sezer Okay, İnci Kazkayasi, Ayşe Filiz Öner
Tuberculosis (TB) is still one of the most common infectious diseases around the world despite the widespread use of BCG (bacille Calmette-Guerin) strain of Mycobacterium bovis as a vaccine. This vaccine does not always protect people from TB, and, thus, new effective vaccines or vaccination strategies are being investigated. In this study, alkyl hydroperoxide reductase (AhpC) from M. bovis was evaluated as a new candidate vaccine antigen against TB in BALB/c mice model. The ahpC gene was amplified from M.bovis genome, cloned, and expressed in Escherichia coli. Vaccine antigen AhpC was formulated with Montanide ISA 61 VG, an oil-based emulsion adjuvant. Both IgG and IL-12 responses were observed in mice after administering the formulation both as a subunit vaccine alone and also as a booster vaccine for BCG immunization. However, a long-lasting response was observed when AhpC formulation was used as a booster (for BCG-primed immunization) as compared to being used as a subunit vaccine alone. In short, these findings suggested that AhpC has the potential to be used as a booster vaccine candidate for BCG-primed immunization.
尽管广泛使用牛分枝杆菌的卡介苗(bacille Calmette-Guerin)菌株作为疫苗,但结核病(TB)仍然是世界上最常见的传染病之一。这种疫苗并不总是保护人们免受结核病的侵害,因此,正在研究新的有效疫苗或疫苗接种策略。本研究在BALB/c小鼠模型中对牛分枝杆菌的烷基氢过氧化物还原酶(AhpC)作为新的结核疫苗候选抗原进行了评价。从牛分枝杆菌基因组中扩增ahpC基因,克隆并在大肠杆菌中表达。用油基乳剂Montanide ISA 61 VG配制疫苗抗原AhpC。在将该制剂单独作为亚单位疫苗和作为卡介苗免疫加强疫苗接种后,在小鼠中观察到IgG和IL-12的反应。然而,与单独作为亚单位疫苗使用相比,将AhpC制剂用作增强剂(用于bcg启动免疫)时观察到持久的应答。总之,这些发现表明AhpC有可能被用作bcg引物免疫的候选加强疫苗。
{"title":"Recombinant AhpC antigen from <i>Mycobacterium bovis</i> boosts BCG-primed immunity in mice.","authors":"Özgün Fırat Düzenli, Sezer Okay, İnci Kazkayasi, Ayşe Filiz Öner","doi":"10.3906/biy-2108-41","DOIUrl":"https://doi.org/10.3906/biy-2108-41","url":null,"abstract":"<p><p>Tuberculosis (TB) is still one of the most common infectious diseases around the world despite the widespread use of BCG (bacille Calmette-Guerin) strain of <i>Mycobacterium bovis</i> as a vaccine. This vaccine does not always protect people from TB, and, thus, new effective vaccines or vaccination strategies are being investigated. In this study, alkyl hydroperoxide reductase (AhpC) from <i>M. bovis</i> was evaluated as a new candidate vaccine antigen against TB in BALB/c mice model. The <i>ahpC</i> gene was amplified from <i>M.bovis</i> genome, cloned, and expressed in <i>Escherichia coli</i>. Vaccine antigen AhpC was formulated with Montanide ISA 61 VG, an oil-based emulsion adjuvant. Both IgG and IL-12 responses were observed in mice after administering the formulation both as a subunit vaccine alone and also as a booster vaccine for BCG immunization. However, a long-lasting response was observed when AhpC formulation was used as a booster (for BCG-primed immunization) as compared to being used as a subunit vaccine alone. In short, these findings suggested that AhpC has the potential to be used as a booster vaccine candidate for BCG-primed immunization.</p>","PeriodicalId":23375,"journal":{"name":"Turkish journal of biology = Turk biyoloji dergisi","volume":"46 1","pages":"95-104"},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10393101/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9923881","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}