Irinotecan (CPT-11), a chemotherapeutic agent used to treat several types of cancer, induces cytotoxic effects on healthy cells. The epidermal growth factor receptor (EGFR) plays a crucial role in various forms of cancer. Nimotuzumab (NmAb), a monoclonal antibody that targets the EGFR, is utilized in some countries to treat malignancies that have an overexpression of EGFR. Yet, there is a lack of literature on the potential anticancer properties of the CPT-11 and NmAb combination on in vitro human cervical cancer cells. This study investigates the apoptosis mode of the CPT-11 and NmAb combination on cervical HeLa cancer cells. The Annexin V/propidium iodide staining examination demonstrated that the combination of CPT-11 and NmAb resulted in a decrease in the number of viable cells and more potent induction of cell apoptosis than the effects of CPT-11 or NmAb alone in HeLa cells. Furthermore, the combined treatment resulted in elevated levels of reactive oxygen species (ROS) and Ca2+ compared to the treatment with CPT-11 or NmAb alone. Cells that were pretreated with N-acetyl-l-cysteine, a substance that scavenges ROS, and then treated with CPT-11, NmAb, or a combination of CPT-11 and NmAb exhibited higher numbers of viable cells compared to those treated with CPT-11 or NmAb alone. The combination of CPT-11 and NmAb resulted in significantly higher caspase-3, -8, and -9 activity levels than CPT-11 or NmAb alone, as measured by flow cytometer assay. The combination of CPT-11 and NmAb in HeLa cells resulted in elevated endoplasmic reticulum stress-, mitochondria-, and caspase-mediated proteins compared to treatment with CPT-11 or NmAb alone. According to these observations, NmAb enhances the effectiveness of CPT-11 in fighting cancer by stimulating cell death in the HeLa cells. Therefore, NmAb has the potential to improve the efficacy of CPT-11 as a future cervical cancer treatment in humans.
{"title":"Nimotuzumab and irinotecan synergistically induce ROS-mediated apoptosis by endoplasmic reticulum stress and mitochondrial-mediated pathway in cervical cancer.","authors":"Fei Teng, Lujun Zhao","doi":"10.1002/bab.2693","DOIUrl":"https://doi.org/10.1002/bab.2693","url":null,"abstract":"<p><p>Irinotecan (CPT-11), a chemotherapeutic agent used to treat several types of cancer, induces cytotoxic effects on healthy cells. The epidermal growth factor receptor (EGFR) plays a crucial role in various forms of cancer. Nimotuzumab (NmAb), a monoclonal antibody that targets the EGFR, is utilized in some countries to treat malignancies that have an overexpression of EGFR. Yet, there is a lack of literature on the potential anticancer properties of the CPT-11 and NmAb combination on in vitro human cervical cancer cells. This study investigates the apoptosis mode of the CPT-11 and NmAb combination on cervical HeLa cancer cells. The Annexin V/propidium iodide staining examination demonstrated that the combination of CPT-11 and NmAb resulted in a decrease in the number of viable cells and more potent induction of cell apoptosis than the effects of CPT-11 or NmAb alone in HeLa cells. Furthermore, the combined treatment resulted in elevated levels of reactive oxygen species (ROS) and Ca<sup>2+</sup> compared to the treatment with CPT-11 or NmAb alone. Cells that were pretreated with N-acetyl-l-cysteine, a substance that scavenges ROS, and then treated with CPT-11, NmAb, or a combination of CPT-11 and NmAb exhibited higher numbers of viable cells compared to those treated with CPT-11 or NmAb alone. The combination of CPT-11 and NmAb resulted in significantly higher caspase-3, -8, and -9 activity levels than CPT-11 or NmAb alone, as measured by flow cytometer assay. The combination of CPT-11 and NmAb in HeLa cells resulted in elevated endoplasmic reticulum stress-, mitochondria-, and caspase-mediated proteins compared to treatment with CPT-11 or NmAb alone. According to these observations, NmAb enhances the effectiveness of CPT-11 in fighting cancer by stimulating cell death in the HeLa cells. Therefore, NmAb has the potential to improve the efficacy of CPT-11 as a future cervical cancer treatment in humans.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142766248","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dysfunction of the alveolar endothelial barrier plays a crucial role in the pathogenesis of septic acute lung injury (ALI). orexin B is a neuropeptide derived from orexin neurons in the lateral hypothalamus and has multiple biological functions. However, the physiological function of orexin B in sepsis is less reported. Here, we aimed to explore the protective effects of orexin B in sepsis-induced ALI and its underlying mechanisms. In this study, we established an ALI in vivo animal model in mice using cecal ligation and puncture (CLP) and an in vitro ALI model using mouse lung microvascular endothelial cells (MLMECs) induced with lipopolysaccharides (LPS). The animal experiments involved four groups: Sham, Sham+orexin B, CLP, CLP+orexin B. First, our results demonstrate that the levels of serum orexin B but not orexin A were reduced in septic mice. Correspondingly, the expression of orexin type 2 receptor (OX2R), but not orexin type 1 receptor (OX1R), was reduced in the lung tissue of septic mice. Administration of orexin B decreased the mortality in sepsis mice and improved M-CASS scores. Hematoxylin-eosin (H&E) staining assay demonstrated that administration of orexin B ameliorated histopathological lung injury. orexin B was also found to inhibit the inflammatory response in the lung tissues of septic mice by reducing the expression of tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and recombinant chemokine C-X-C-motif ligand 15 (CXCL15). Additionally, the total cell count and neutrophils in bronchoalveolar lavage fluid (BALF) were reduced by orexin B. Notably, orexin B alleviated vascular endothelial permeability in mice lung tissue by increasing the expression of the tight junction protein zonula occludens-1 (ZO-1) and occludin. In vitro experiments demonstrated that orexin B prevented LPS-induced endothelial permeability in mouse lung microvascular endothelial cells (MLMECs) by upregulating the expression of ZO-1 and occludin. These effects are mediated by rho-associated coiled-coil containing protein kinase 2 (ROCK2). Based on these findings, we conclude that orexin B alleviates sepsis-induced ALI by ameliorating endothelial permeability of lung microvascular endothelial cells.
{"title":"orexin B alleviates sepsis-associated lung injury through the attenuation of pulmonary endothelial barrier dysfunction by regulating the rho-associated coiled-coil containing protein kinase 2/zonula occludens-1 (ROCK2/ZO-1) axis.","authors":"Yiyuan Wang, Xiaohong Wan, Yusheng Li","doi":"10.1002/bab.2703","DOIUrl":"https://doi.org/10.1002/bab.2703","url":null,"abstract":"<p><p>Dysfunction of the alveolar endothelial barrier plays a crucial role in the pathogenesis of septic acute lung injury (ALI). orexin B is a neuropeptide derived from orexin neurons in the lateral hypothalamus and has multiple biological functions. However, the physiological function of orexin B in sepsis is less reported. Here, we aimed to explore the protective effects of orexin B in sepsis-induced ALI and its underlying mechanisms. In this study, we established an ALI in vivo animal model in mice using cecal ligation and puncture (CLP) and an in vitro ALI model using mouse lung microvascular endothelial cells (MLMECs) induced with lipopolysaccharides (LPS). The animal experiments involved four groups: Sham, Sham+orexin B, CLP, CLP+orexin B. First, our results demonstrate that the levels of serum orexin B but not orexin A were reduced in septic mice. Correspondingly, the expression of orexin type 2 receptor (OX2R), but not orexin type 1 receptor (OX1R), was reduced in the lung tissue of septic mice. Administration of orexin B decreased the mortality in sepsis mice and improved M-CASS scores. Hematoxylin-eosin (H&E) staining assay demonstrated that administration of orexin B ameliorated histopathological lung injury. orexin B was also found to inhibit the inflammatory response in the lung tissues of septic mice by reducing the expression of tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and recombinant chemokine C-X-C-motif ligand 15 (CXCL15). Additionally, the total cell count and neutrophils in bronchoalveolar lavage fluid (BALF) were reduced by orexin B. Notably, orexin B alleviated vascular endothelial permeability in mice lung tissue by increasing the expression of the tight junction protein zonula occludens-1 (ZO-1) and occludin. In vitro experiments demonstrated that orexin B prevented LPS-induced endothelial permeability in mouse lung microvascular endothelial cells (MLMECs) by upregulating the expression of ZO-1 and occludin. These effects are mediated by rho-associated coiled-coil containing protein kinase 2 (ROCK2). Based on these findings, we conclude that orexin B alleviates sepsis-induced ALI by ameliorating endothelial permeability of lung microvascular endothelial cells.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142766251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiajie Zhang, Ming Qu, Zhanhao Mo, He Sui, Lin Liu, Deliang Fu
In prior research, both miRNA-125b and BLZ945 have shown potential in effectively inhibiting M2 macrophage polarization and producing antitumor effects. Nevertheless, their physicochemical characteristics present significant challenges for efficient in vivo delivery. Ionizable cationic lipid nanoparticles (LNPs), recognized for their superior biocompatibility and drug-loading capacity, serve as a novel carrier for nucleic acid-based therapeutics. In our study, we successfully encapsulated both agents within LNPs and conducted a thorough characterization. Subsequently, we investigated their potential to repolarize M2 macrophages in vitro and evaluated their in vivo distribution, biosafety, and antitumor efficacy. The findings revealed that the LNPs maintained excellent drug-loading efficiency, consistent particle size, and stable zeta potential. All formulations effectively inhibited M2 macrophage polarization in vitro. Upon administration in vivo, the LNPs not only demonstrated favorable biosafety profiles but also accumulated efficiently in tumor tissues, substantially reducing tumor burden, particularly notable in co-loaded LNPs. Our results affirm that LNPs are an effective carrier for miRNA-125b and BLZ945, highlighting this encapsulation approach as promising for the treatment of solid tumors and meriting further investigation. Practitioner points: (i) Ionizable cationic nanoparticles provide high and stable encapsulation rates to efficiently load nucleic acid polymers into the LNP, avoiding the rapid accumulation of circulating macrophages, which can lead to reduced penetration of the LNP into target tissues. Therefore, it can be used as a novel drug delivery method to benefit clinical patients. (ii) miRNA-125b LNP/BLZ945 LNP attenuated the depleting effect of BLZ945 on macrophages and significantly inhibited macrophage M2 polarization. It could be effectively distributed in tumors and showed good biosafety while exerting antitumor effects, bringing hope to clinical pancreatic tumor patients.
{"title":"Ionizable cationic lipid nanoparticles loaded with miRNA-125b/BLZ945 for pancreatic cancer treatment.","authors":"Jiajie Zhang, Ming Qu, Zhanhao Mo, He Sui, Lin Liu, Deliang Fu","doi":"10.1002/bab.2701","DOIUrl":"https://doi.org/10.1002/bab.2701","url":null,"abstract":"<p><p>In prior research, both miRNA-125b and BLZ945 have shown potential in effectively inhibiting M2 macrophage polarization and producing antitumor effects. Nevertheless, their physicochemical characteristics present significant challenges for efficient in vivo delivery. Ionizable cationic lipid nanoparticles (LNPs), recognized for their superior biocompatibility and drug-loading capacity, serve as a novel carrier for nucleic acid-based therapeutics. In our study, we successfully encapsulated both agents within LNPs and conducted a thorough characterization. Subsequently, we investigated their potential to repolarize M2 macrophages in vitro and evaluated their in vivo distribution, biosafety, and antitumor efficacy. The findings revealed that the LNPs maintained excellent drug-loading efficiency, consistent particle size, and stable zeta potential. All formulations effectively inhibited M2 macrophage polarization in vitro. Upon administration in vivo, the LNPs not only demonstrated favorable biosafety profiles but also accumulated efficiently in tumor tissues, substantially reducing tumor burden, particularly notable in co-loaded LNPs. Our results affirm that LNPs are an effective carrier for miRNA-125b and BLZ945, highlighting this encapsulation approach as promising for the treatment of solid tumors and meriting further investigation. Practitioner points: (i) Ionizable cationic nanoparticles provide high and stable encapsulation rates to efficiently load nucleic acid polymers into the LNP, avoiding the rapid accumulation of circulating macrophages, which can lead to reduced penetration of the LNP into target tissues. Therefore, it can be used as a novel drug delivery method to benefit clinical patients. (ii) miRNA-125b LNP/BLZ945 LNP attenuated the depleting effect of BLZ945 on macrophages and significantly inhibited macrophage M2 polarization. It could be effectively distributed in tumors and showed good biosafety while exerting antitumor effects, bringing hope to clinical pancreatic tumor patients.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142766235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Metal-based nanoparticles have been extensively researched for their distinctive characteristics. Among them, zinc oxide nanoparticles have numerous applications in the field of biomedicine. The phytoextract of Ixora coccinea flowers was used in the synthesis of ZnO nanoparticles replacing the use of harmful reducing chemicals. In the current research, the carbonaceous material from biowaste of Setaria italica was used to synthesize graphene oxide (GO) by Improved Hummer's method. The synthesized GO was converted to reduced GO via green nanotechnology using phytoextract of Prosopis juliflora. The synthesis of reduced Graphene Oxide - Zinc Oxide Nanocomposite (rGO)-ZnO nanocomposite involves a simple, economical one-step magnetic stirring method. UV-visible spectroscopy was used to characterize the synthesized materials, with the maximal absorbance range for ZInc Oxide (ZnO) being 384 nm and for rGO-ZnO composite at 243 and 366 nm, respectively. The x-ray diffraction (XRD) revealed 2θ peaks for ZnO at 31.54°, 34.22°, and 36.08°. For reduced Graphene Oxide (rGO) in rGO-ZnO composite, the XRD revealed 2θ peaks at 21.25°, 21.56°, 23.14°, and for ZnO at 31.74°, 33.24°, 34.29°, 36.23°. The FT-IR demonstrated the vibrational modes of functional groups: -OH stretching, symmetric and antisymmetric -CH2 stretching, C = C stretching, and C-O stretching. The elemental composition of samples has been analyzed using Energy Dispersive x-ray spectroscop (EDX), and the high percentage of zinc in the composite shows a good loading rate of ZnO on the rGO's surface. By morphological investigation, monolayer sheet structures of rGO loaded with clusters of ZnO are clearly demonstrated. Positive results from therapeutic assays and biocompatibility were found with reduced hemolysis and good anticoagulation abilities proved with statistical approach. Our research is distinctive because a realistic formulation of an rGO-ZnO skin care cream with enhanced therapeutic properties, such as effective stability, spreadability, and significant moisture retention, can be recommended.
{"title":"Sustainable strategy of biowaste into graphene-based zinc oxide nanocomposite using green nanotechnology for topical applications.","authors":"Chamundeeswari M, Preethy Kr","doi":"10.1002/bab.2702","DOIUrl":"https://doi.org/10.1002/bab.2702","url":null,"abstract":"<p><p>Metal-based nanoparticles have been extensively researched for their distinctive characteristics. Among them, zinc oxide nanoparticles have numerous applications in the field of biomedicine. The phytoextract of Ixora coccinea flowers was used in the synthesis of ZnO nanoparticles replacing the use of harmful reducing chemicals. In the current research, the carbonaceous material from biowaste of Setaria italica was used to synthesize graphene oxide (GO) by Improved Hummer's method. The synthesized GO was converted to reduced GO via green nanotechnology using phytoextract of Prosopis juliflora. The synthesis of reduced Graphene Oxide - Zinc Oxide Nanocomposite (rGO)-ZnO nanocomposite involves a simple, economical one-step magnetic stirring method. UV-visible spectroscopy was used to characterize the synthesized materials, with the maximal absorbance range for ZInc Oxide (ZnO) being 384 nm and for rGO-ZnO composite at 243 and 366 nm, respectively. The x-ray diffraction (XRD) revealed 2θ peaks for ZnO at 31.54°, 34.22°, and 36.08°. For reduced Graphene Oxide (rGO) in rGO-ZnO composite, the XRD revealed 2θ peaks at 21.25°, 21.56°, 23.14°, and for ZnO at 31.74°, 33.24°, 34.29°, 36.23°. The FT-IR demonstrated the vibrational modes of functional groups: -OH stretching, symmetric and antisymmetric -CH<sub>2</sub> stretching, C = C stretching, and C-O stretching. The elemental composition of samples has been analyzed using Energy Dispersive x-ray spectroscop (EDX), and the high percentage of zinc in the composite shows a good loading rate of ZnO on the rGO's surface. By morphological investigation, monolayer sheet structures of rGO loaded with clusters of ZnO are clearly demonstrated. Positive results from therapeutic assays and biocompatibility were found with reduced hemolysis and good anticoagulation abilities proved with statistical approach. Our research is distinctive because a realistic formulation of an rGO-ZnO skin care cream with enhanced therapeutic properties, such as effective stability, spreadability, and significant moisture retention, can be recommended.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142766273","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dilek Sönmezer Açıkgöz, Fatma Latifoğlu, Güler Toprak, Münevver Baran
Enhancing the biocompatibility of biomaterials is a critical aspect of tissue engineering and regenerative medicine. Advances in 3D bioprinting technology, blending natural and synthetic materials for the production of bioink, offer new opportunities to develop highly biocompatible materials that can closely mimic the native tissue environment. In this study, we used pericardial fluid structure (PFS)-based material together with alginate to mimic the extracellular matrix (ECM) and produce a bioink material. Thus, blended alginate with PFS material and MC3T3-E1 pre-osteoblast cell-laden hydrogels characterized by comparing each other, especially alginate hydrogels, and evaluated in terms of biocompatibility for tissue engineering applications. According to the rheological analysis results, all hydrogel groups A, A-PFS (150 mg), and A-PFS (1:1) had viscoelastic properties. Mechanical tests showed that the A-PFS (1:1) hydrogel had the most strength properties. Additionally, the viscosity values of the hydrogel solutions were in an applicable range for use in 3D bioprinters. It was also found out that PFS increased the biocompatibility of alginate-based bioink, in terms of cell proliferation and differentiation. Overall, these findings suggest that alginate and pericardial fluid-based materials can be successfully used for bioink production. The resulting hydrogels exhibit viscoelastic properties, appropriate viscosity for 3D bioprinting, and support cell viability, proliferation, and osteogenic differentiation. This research has the potential not only to produce bioink but also to produce injectable hydrogels and drug delivery systems, which can become biocompatible materials that can be used for tissue engineering and regenerative medicine applications.
{"title":"Applications of a biocompatible alginate/pericardial fluid-based hydrogel for the production of a bioink in tissue engineering.","authors":"Dilek Sönmezer Açıkgöz, Fatma Latifoğlu, Güler Toprak, Münevver Baran","doi":"10.1002/bab.2697","DOIUrl":"https://doi.org/10.1002/bab.2697","url":null,"abstract":"<p><p>Enhancing the biocompatibility of biomaterials is a critical aspect of tissue engineering and regenerative medicine. Advances in 3D bioprinting technology, blending natural and synthetic materials for the production of bioink, offer new opportunities to develop highly biocompatible materials that can closely mimic the native tissue environment. In this study, we used pericardial fluid structure (PFS)-based material together with alginate to mimic the extracellular matrix (ECM) and produce a bioink material. Thus, blended alginate with PFS material and MC3T3-E1 pre-osteoblast cell-laden hydrogels characterized by comparing each other, especially alginate hydrogels, and evaluated in terms of biocompatibility for tissue engineering applications. According to the rheological analysis results, all hydrogel groups A, A-PFS (150 mg), and A-PFS (1:1) had viscoelastic properties. Mechanical tests showed that the A-PFS (1:1) hydrogel had the most strength properties. Additionally, the viscosity values of the hydrogel solutions were in an applicable range for use in 3D bioprinters. It was also found out that PFS increased the biocompatibility of alginate-based bioink, in terms of cell proliferation and differentiation. Overall, these findings suggest that alginate and pericardial fluid-based materials can be successfully used for bioink production. The resulting hydrogels exhibit viscoelastic properties, appropriate viscosity for 3D bioprinting, and support cell viability, proliferation, and osteogenic differentiation. This research has the potential not only to produce bioink but also to produce injectable hydrogels and drug delivery systems, which can become biocompatible materials that can be used for tissue engineering and regenerative medicine applications.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142765875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ayca Zeybek Kuyucu, Dogu Sayili, Ridvan Orkut, Olcay Mert, İbrahim Oguzhan Tarman, Busra Lulaci, Ali Mert Sencer, Asli Kurden Pekmezci, Mehmet Ender Avci, Sibel Kalyoncu, Mehmet Inan
Checkpoint inhibitors are widely recognized immunotherapeutic drugs, known for their effectiveness in treating various cancers. Atezolizumab, targeting the immune checkpoint programmed death-ligand 1, is successfully used to treat several types of cancers. Atezolizumab is a potential biosimilar candidate due to its huge success in the clinic but there is no literature on its production process in mammalian cells. In this study, we generated a monoclonal cell line derived from recombinant Chinese hamster ovary DG44 cells to produce atezolizumab. The selected single clone was employed for media screening and process development. Following production in a 7-L bioreactor, atezolizumab was purified using a three-step chromatographic method. Finally, the purified atezolizumab was characterized and compared with commercial atezolizumab (Tecentriq) through several chromatographic and kinetics analyses.
{"title":"Cell line development and bioreactor process optimization for an atezolizumab biosimilar.","authors":"Ayca Zeybek Kuyucu, Dogu Sayili, Ridvan Orkut, Olcay Mert, İbrahim Oguzhan Tarman, Busra Lulaci, Ali Mert Sencer, Asli Kurden Pekmezci, Mehmet Ender Avci, Sibel Kalyoncu, Mehmet Inan","doi":"10.1002/bab.2704","DOIUrl":"https://doi.org/10.1002/bab.2704","url":null,"abstract":"<p><p>Checkpoint inhibitors are widely recognized immunotherapeutic drugs, known for their effectiveness in treating various cancers. Atezolizumab, targeting the immune checkpoint programmed death-ligand 1, is successfully used to treat several types of cancers. Atezolizumab is a potential biosimilar candidate due to its huge success in the clinic but there is no literature on its production process in mammalian cells. In this study, we generated a monoclonal cell line derived from recombinant Chinese hamster ovary DG44 cells to produce atezolizumab. The selected single clone was employed for media screening and process development. Following production in a 7-L bioreactor, atezolizumab was purified using a three-step chromatographic method. Finally, the purified atezolizumab was characterized and compared with commercial atezolizumab (Tecentriq) through several chromatographic and kinetics analyses.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142766182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Triple-negative breast cancer (TNBC), known for its hostile nature and limited treatment modalities, has spurred researchers to explore novel approaches for enhancing clinical outcomes. Here, the study aimed to analyze transcriptomics data to identify immune-related hub genes associated with TNBC that might serve as prognostic biomarkers. Initially, we determined genes that were differentially expressed between TNBC and normal tissues by integrating microarray and RNA sequencing data. Then, through protein-protein interaction and module analysis, we identified five putative hub genes: AURKA, CCNB1, CDCA8, GAPDH, and TOP2A. Subsequently, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that the hub genes were primarily involved in the progesterone-mediated oocyte maturation signaling pathway and oocyte meiosis. Additionally, we observed that these five hub genes were significantly elevated at both protein and mRNA levels in TNBC tissues and contributed to worse survival. Furthermore, the expression of these hub genes exhibited a strong positive association with immune-invading cells such as CD8 T cells, CD4 T cells, and dendritic cells. The analysis of the regulatory network revealed three transcription factors (YBX-1, E2F1, and E2F3) and three posttranscriptional regulators (hsa-mir-25-3p, hsa-mir-92a-3p, and hsa-let-7b-5p) of hub genes. Finally, we explored potential drug candidates for the hub genes using Drug-Gene Interaction Database and discovered that there are no FDA-approved drugs for CCNB1 and CDCA8, highlighting a promising area for future research. Taken together, our results will be of immense importance in addressing the intricacies of TNBC.
{"title":"Deciphering the prognostic landscape of triple-negative breast cancer: A focus on immune-related hub genes and therapeutic implications.","authors":"HemaNandini Rajendran Krishnamoorthy, Ramanathan Karuppasamy","doi":"10.1002/bab.2700","DOIUrl":"https://doi.org/10.1002/bab.2700","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC), known for its hostile nature and limited treatment modalities, has spurred researchers to explore novel approaches for enhancing clinical outcomes. Here, the study aimed to analyze transcriptomics data to identify immune-related hub genes associated with TNBC that might serve as prognostic biomarkers. Initially, we determined genes that were differentially expressed between TNBC and normal tissues by integrating microarray and RNA sequencing data. Then, through protein-protein interaction and module analysis, we identified five putative hub genes: AURKA, CCNB1, CDCA8, GAPDH, and TOP2A. Subsequently, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that the hub genes were primarily involved in the progesterone-mediated oocyte maturation signaling pathway and oocyte meiosis. Additionally, we observed that these five hub genes were significantly elevated at both protein and mRNA levels in TNBC tissues and contributed to worse survival. Furthermore, the expression of these hub genes exhibited a strong positive association with immune-invading cells such as CD8 T cells, CD4 T cells, and dendritic cells. The analysis of the regulatory network revealed three transcription factors (YBX-1, E2F1, and E2F3) and three posttranscriptional regulators (hsa-mir-25-3p, hsa-mir-92a-3p, and hsa-let-7b-5p) of hub genes. Finally, we explored potential drug candidates for the hub genes using Drug-Gene Interaction Database and discovered that there are no FDA-approved drugs for CCNB1 and CDCA8, highlighting a promising area for future research. Taken together, our results will be of immense importance in addressing the intricacies of TNBC.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715442","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Vasilisa V Krasitskaya, Kristina A Drandrova, Anna V Tyumentseva, Irina G Vazhenina, Anna V Lukyanenko, Sergey V Stolyar, Ludmila A Frank
Exosomes are a type of membrane vesicle secreted into the extracellular medium by most cell types. They have a great potential for clinical practice as noninvasive biomarkers for diagnosis of various diseases, prognosis, and monitoring of therapy, which stimulates the development of simple methods for isolating exosomes from biological fluids. A novel affine material based on aminosilanized superparamagnetic core‒shell nanoparticles for fast isolation of urinary exosomes is reported. Iron oxide nanoparticles coated with amino organosilane have been synthesized. The structural and magnetic characteristics of the resulting nanoparticles have been studied by transmission electron microscopy and ferromagnetic resonance. The surface of the synthesized nanoparticles has been chemically functionalized with lectin (concanavalin A), and the efficiency of the obtained material as a sorbent for affine exosome isolation from human urine has been demonstrated. A highly purified fraction of exosomes 90-200 nm in size has been obtained. The exosomal nature of the isolated vesicles has been confirmed by bioluminescent solid-phase microassay of tetrasporine receptor markers. The presence of exosomal miR-21 in the isolated human urine exosome samples has been established.
{"title":"Concanavalin A-activated magnetic nanoparticles as an affine material for urinary exosome isolation.","authors":"Vasilisa V Krasitskaya, Kristina A Drandrova, Anna V Tyumentseva, Irina G Vazhenina, Anna V Lukyanenko, Sergey V Stolyar, Ludmila A Frank","doi":"10.1002/bab.2696","DOIUrl":"https://doi.org/10.1002/bab.2696","url":null,"abstract":"<p><p>Exosomes are a type of membrane vesicle secreted into the extracellular medium by most cell types. They have a great potential for clinical practice as noninvasive biomarkers for diagnosis of various diseases, prognosis, and monitoring of therapy, which stimulates the development of simple methods for isolating exosomes from biological fluids. A novel affine material based on aminosilanized superparamagnetic core‒shell nanoparticles for fast isolation of urinary exosomes is reported. Iron oxide nanoparticles coated with amino organosilane have been synthesized. The structural and magnetic characteristics of the resulting nanoparticles have been studied by transmission electron microscopy and ferromagnetic resonance. The surface of the synthesized nanoparticles has been chemically functionalized with lectin (concanavalin A), and the efficiency of the obtained material as a sorbent for affine exosome isolation from human urine has been demonstrated. A highly purified fraction of exosomes 90-200 nm in size has been obtained. The exosomal nature of the isolated vesicles has been confirmed by bioluminescent solid-phase microassay of tetrasporine receptor markers. The presence of exosomal miR-21 in the isolated human urine exosome samples has been established.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686064","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Osteoarthritis (OA) is a degenerative joint disorder characterized by progressive cartilage degradation. Excessive oxidative stress (OS), inflammatory responses, extracellular matrix breakdown, and cellular senescence of chondrocytes play crucial roles in the pathological development of OA. Currently, curing OA remains a significant challenge. In this study, we aimed to elucidate the protective effects of Annexin A1 protein Mimetic Peptide (Ac2-26) against tumor necrosis factor-α (TNF-α)-induced damage in CHON-001 chondrocytes by assessing cellular senescence, OS, and the expression levels of matrix metalloproteinase-13 (MMP-13) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4. Our results show that Ac2-26 mitigated the reduction of telomerase activity and the exacerbation of cellular senescence induced by TNF-α in CHON-001 chondrocytes. Treatment with TNF-α led to decreased expression of the human telomerase reverse transcriptase gene and increased expression of the telomeric repeat-binding factor 2 gene, which were reversed by Ac2-26 treatment. The TNF-α-induced increases in the gene and protein expressions of p53 and p16 were restored by Ac2-26 in a dose-dependent manner. Additionally, we found that TNF-α caused elevations in the mRNA and protein levels of MMP-13 and ADAMTS-4, which were reduced by Ac2-26 in a dose-dependent fashion. Furthermore, TNF-α triggered the activation of nuclear factor κ-B (NF-κB) by increasing the levels of phosphorylated NF-κB p65 and the luciferase activity of NF-κB. Notably, Ac2-26 alleviated OS by reducing mitochondrial reactive oxygen species levels and promoting the activation of NF-E2-related factor 2 (Nrf2) in TNF-α-challenged CHON-001 chondrocytes. Silencing Nrf2 abolished the Ac2-26-induced activation of NF-κB and cellular senescence in CHON-001 chondrocytes. Collectively, these findings offer new insights into the potential therapeutic use of Ac2-26 for treating OA.
{"title":"The Annexin A1 Protein Mimetic Peptide Ac2-26 prevents cellular senescence of CHON-001 chondrocytes against tumor necrosis factor-α via the Nrf2/NF-κB pathway.","authors":"Lei Yang, Kaijian Gong, Guoxing Ren, Bo Chen","doi":"10.1002/bab.2695","DOIUrl":"https://doi.org/10.1002/bab.2695","url":null,"abstract":"<p><p>Osteoarthritis (OA) is a degenerative joint disorder characterized by progressive cartilage degradation. Excessive oxidative stress (OS), inflammatory responses, extracellular matrix breakdown, and cellular senescence of chondrocytes play crucial roles in the pathological development of OA. Currently, curing OA remains a significant challenge. In this study, we aimed to elucidate the protective effects of Annexin A1 protein Mimetic Peptide (Ac2-26) against tumor necrosis factor-α (TNF-α)-induced damage in CHON-001 chondrocytes by assessing cellular senescence, OS, and the expression levels of matrix metalloproteinase-13 (MMP-13) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4. Our results show that Ac2-26 mitigated the reduction of telomerase activity and the exacerbation of cellular senescence induced by TNF-α in CHON-001 chondrocytes. Treatment with TNF-α led to decreased expression of the human telomerase reverse transcriptase gene and increased expression of the telomeric repeat-binding factor 2 gene, which were reversed by Ac2-26 treatment. The TNF-α-induced increases in the gene and protein expressions of p53 and p16 were restored by Ac2-26 in a dose-dependent manner. Additionally, we found that TNF-α caused elevations in the mRNA and protein levels of MMP-13 and ADAMTS-4, which were reduced by Ac2-26 in a dose-dependent fashion. Furthermore, TNF-α triggered the activation of nuclear factor κ-B (NF-κB) by increasing the levels of phosphorylated NF-κB p65 and the luciferase activity of NF-κB. Notably, Ac2-26 alleviated OS by reducing mitochondrial reactive oxygen species levels and promoting the activation of NF-E2-related factor 2 (Nrf2) in TNF-α-challenged CHON-001 chondrocytes. Silencing Nrf2 abolished the Ac2-26-induced activation of NF-κB and cellular senescence in CHON-001 chondrocytes. Collectively, these findings offer new insights into the potential therapeutic use of Ac2-26 for treating OA.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686067","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cisplatin, the pioneering heavy metal compound, stands out as a potent drug for the treatment of various solid tumors. However, its clinical utility is hampered by notable toxicity and adverse effects, particularly nephrotoxicity. The potency of rutecarpine, a phytochemical, in mitigating cisplatin-induced nephrotoxicity was assessed in the present study. In this experimental setup, healthy male Wistar rats were grouped into four and Group I rats served as the control group, receiving only vehicle control. Group II rats were subjected to cisplatin treatment alone, administered intraperitoneally at a dosage of 7 mg/kg body weight on the 19th, 20th, and 21st days. Group III and IV rats were orally administered with rutecarpine at doses of 10 and 20 mg/kg body weight, respectively, starting from Day 1 and continuing daily for 21 days. Additionally, they were injected intraperitoneally with cisplatin at the same dosage and schedule as Group II. Relative kidney weight and renal biochemical markers blood urea nitrogen, lactate dehydrogenase, serum urea, and creatinine were measured to assess rutecarpine inhibitory potency against cisplatin toxicity. Markers of oxidative damage and antioxidants levels were quantified in the ruteacarpine- and cisplatin-treated rats. The study investigated the anti-inflammatory property of rutecarpine in cisplatin-induced nephrotoxicity by analyzing inflammatory cytokines. Renal tissue levels of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B, key markers of nephrotoxicity, were quantified to assess rutecarpine's potential to mitigate cisplatin-triggered damage. Histopathological examinations were performed to confirm the impact of rutecarpine against cisplatin-induced nephrotoxicity. Treatment with rutecarpine notably reduced renal biochemical markers, prevented renal edema, and attenuated oxidative stress-induced damage in cisplatin-treated rats. Both inflammatory and nephrotoxicity markers showed significant decreases in rats treated with rutecarpine along with cisplatin. Histological analysis affirmed that rutecarpine pretreatment effectively prevented cisplatin-induced nephrotoxicity. The study findings demonstrate that rutecarpine ameliorates cisplatin-triggered nephrotoxicity through its antioxidant and anti-inflammatory properties, suggesting that rutecarpine supplementation alongside cisplatin treatment could potentially reduce nephrotoxicity in cancer patients.
{"title":"Ameliorative effect of rutecarpine supplementation against cisplatin-induced nephrotoxicity in rats via inhibition of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B.","authors":"Dong Zhang, Rui Jin, Guoxing Li, CaiFeng Zhang, Yanhong Zhou","doi":"10.1002/bab.2692","DOIUrl":"10.1002/bab.2692","url":null,"abstract":"<p><p>Cisplatin, the pioneering heavy metal compound, stands out as a potent drug for the treatment of various solid tumors. However, its clinical utility is hampered by notable toxicity and adverse effects, particularly nephrotoxicity. The potency of rutecarpine, a phytochemical, in mitigating cisplatin-induced nephrotoxicity was assessed in the present study. In this experimental setup, healthy male Wistar rats were grouped into four and Group I rats served as the control group, receiving only vehicle control. Group II rats were subjected to cisplatin treatment alone, administered intraperitoneally at a dosage of 7 mg/kg body weight on the 19th, 20th, and 21st days. Group III and IV rats were orally administered with rutecarpine at doses of 10 and 20 mg/kg body weight, respectively, starting from Day 1 and continuing daily for 21 days. Additionally, they were injected intraperitoneally with cisplatin at the same dosage and schedule as Group II. Relative kidney weight and renal biochemical markers blood urea nitrogen, lactate dehydrogenase, serum urea, and creatinine were measured to assess rutecarpine inhibitory potency against cisplatin toxicity. Markers of oxidative damage and antioxidants levels were quantified in the ruteacarpine- and cisplatin-treated rats. The study investigated the anti-inflammatory property of rutecarpine in cisplatin-induced nephrotoxicity by analyzing inflammatory cytokines. Renal tissue levels of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B, key markers of nephrotoxicity, were quantified to assess rutecarpine's potential to mitigate cisplatin-triggered damage. Histopathological examinations were performed to confirm the impact of rutecarpine against cisplatin-induced nephrotoxicity. Treatment with rutecarpine notably reduced renal biochemical markers, prevented renal edema, and attenuated oxidative stress-induced damage in cisplatin-treated rats. Both inflammatory and nephrotoxicity markers showed significant decreases in rats treated with rutecarpine along with cisplatin. Histological analysis affirmed that rutecarpine pretreatment effectively prevented cisplatin-induced nephrotoxicity. The study findings demonstrate that rutecarpine ameliorates cisplatin-triggered nephrotoxicity through its antioxidant and anti-inflammatory properties, suggesting that rutecarpine supplementation alongside cisplatin treatment could potentially reduce nephrotoxicity in cancer patients.</p>","PeriodicalId":9274,"journal":{"name":"Biotechnology and applied biochemistry","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142590182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}