Although the number of breast cancer deaths has decreased, and there have been developments in targeted therapies and combination treatments for the management of metastatic illness, metastatic breast cancer is still the second most common cause of cancer-related deaths in U.S. women. Numerous phases and a vast number of proteins and signaling molecules are involved in the invasion-metastasis cascade. The tumor cells penetrate and enter the blood or lymphatic vessels, and travel to distant organs via the lymphatic or blood vessels. Tumor cells enter cell cycle arrest, adhere to capillary beds in the target organ, and then disseminate throughout the organ's parenchyma, proliferating and enhancing angiogenesis. Each of these processes is regulated by changes in the expression of different genes, in which lncRNAs play a role in this regulation. Transcripts that are longer than 200 nucleotides and do not translate into proteins are called RNAs. LncRNA molecules, whose function depends on their unique molecular structure, play significant roles in controlling the expression of genes at various epigenetic levels, transcription, and so on. LncRNAs have essential functions in regulating the expression of genes linked to cell development in healthy and pathological processes, specialization, programmed cell death, cell division, invasion, DNA damage, and spread to other parts of the body. A number of cancer types have been shown to exhibit aberrant expression of lncRNAs. In this review, we describe the general characteristics, potential molecular mechanisms and targeted therapy of lncRNAs and discuss the emerging functions of lncRNAs in breast cancer.
{"title":"Molecular Mechanism of lncRNAs in Regulation of Breast Cancer Metastasis; a Comprehensive Review.","authors":"Shokoufeh Ahmadi, Farzaneh Yazdi, Sahar Khastar, Irwanjot Kaur, Mareb Hamed Ahmed, Abhishek Kumar, Gulshan Rathore, Parjinder Kaur, Mohammad Shahsavan, Mahmoud Dehghani-Ghorbi, Reza Akhavan-Sigari","doi":"10.1007/s12013-024-01535-y","DOIUrl":"https://doi.org/10.1007/s12013-024-01535-y","url":null,"abstract":"<p><p>Although the number of breast cancer deaths has decreased, and there have been developments in targeted therapies and combination treatments for the management of metastatic illness, metastatic breast cancer is still the second most common cause of cancer-related deaths in U.S. women. Numerous phases and a vast number of proteins and signaling molecules are involved in the invasion-metastasis cascade. The tumor cells penetrate and enter the blood or lymphatic vessels, and travel to distant organs via the lymphatic or blood vessels. Tumor cells enter cell cycle arrest, adhere to capillary beds in the target organ, and then disseminate throughout the organ's parenchyma, proliferating and enhancing angiogenesis. Each of these processes is regulated by changes in the expression of different genes, in which lncRNAs play a role in this regulation. Transcripts that are longer than 200 nucleotides and do not translate into proteins are called RNAs. LncRNA molecules, whose function depends on their unique molecular structure, play significant roles in controlling the expression of genes at various epigenetic levels, transcription, and so on. LncRNAs have essential functions in regulating the expression of genes linked to cell development in healthy and pathological processes, specialization, programmed cell death, cell division, invasion, DNA damage, and spread to other parts of the body. A number of cancer types have been shown to exhibit aberrant expression of lncRNAs. In this review, we describe the general characteristics, potential molecular mechanisms and targeted therapy of lncRNAs and discuss the emerging functions of lncRNAs in breast cancer.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142374943","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}
Pub Date : 2024-10-04DOI: 10.1007/s12013-024-01556-7
Yi Tang, Ye Tian, Chun-Xia Zhang, Guo-Tai Wang
Olfactory receptors (ORs) are a class of G protein-coupled receptors (GPCR) widely distributed in olfactory sensory neurons and various non-olfactory tissues, serving significant physiological and pathological functions in the human body. Increasing evidence reveals the heightened expression of olfactory receptors in tumorous tissues and cells alongside normal tissues. Olfactory receptors have demonstrated influence over tumor cell proliferation and metastasis, establishing a close relationship with tumor initiation and progression. This review highlights the specific molecular actions and signaling pathways of olfactory receptors in the development of human tumors. The potential for precise tumor diagnosis and targeted therapy through therapeutic targeting of olfactory receptors as an adjunct anticancer treatment strategy is being considered.
嗅觉受体(ORs)是一类 G 蛋白偶联受体(GPCR),广泛分布于嗅觉神经元和各种非嗅觉组织中,在人体内发挥着重要的生理和病理功能。越来越多的证据表明,嗅觉受体在肿瘤组织和细胞中的表达与正常组织相同。嗅觉受体对肿瘤细胞的增殖和转移有影响,与肿瘤的发生和发展密切相关。本综述将重点介绍嗅觉受体在人类肿瘤发生发展过程中的特定分子作用和信号通路。通过将嗅觉受体作为辅助抗癌治疗策略的治疗靶点,为肿瘤的精确诊断和靶向治疗提供了可能性。
{"title":"Olfactory Receptors and Tumorigenesis: Implications for Diagnosis and Targeted Therapy.","authors":"Yi Tang, Ye Tian, Chun-Xia Zhang, Guo-Tai Wang","doi":"10.1007/s12013-024-01556-7","DOIUrl":"https://doi.org/10.1007/s12013-024-01556-7","url":null,"abstract":"<p><p>Olfactory receptors (ORs) are a class of G protein-coupled receptors (GPCR) widely distributed in olfactory sensory neurons and various non-olfactory tissues, serving significant physiological and pathological functions in the human body. Increasing evidence reveals the heightened expression of olfactory receptors in tumorous tissues and cells alongside normal tissues. Olfactory receptors have demonstrated influence over tumor cell proliferation and metastasis, establishing a close relationship with tumor initiation and progression. This review highlights the specific molecular actions and signaling pathways of olfactory receptors in the development of human tumors. The potential for precise tumor diagnosis and targeted therapy through therapeutic targeting of olfactory receptors as an adjunct anticancer treatment strategy is being considered.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370580","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}
Pub Date : 2024-10-04DOI: 10.1007/s12013-024-01573-6
Tineng Yang, Xiaoyang Peng, Xi Huang, Peng Cao, Hualei Chen
Bladder cancer (BLCA) is a prevalent malignancy worldwide with a high recurrence rate. Collagen Type VI Alpha 1 (COL6A1) plays a key role in several cancer types. In this study, we aimed to explore the role of COL6A1 in BLCA. COL6A1 expression in BLCA was determined using The Cancer Genome Atlas database and real-time quantitative polymerase chain reaction (RT-qPCR). Counting Kit-8, wound-healing, and transwell assays were used to assess the effect of COL6A1 on T24 and 5637 cells. Apoptosis in BLCA cell lines was explored using western blotting and flow cytometry. Co-immunoprecipitation was performed to determine interactions between proteins. The role of COL6A1 in tumor growth in nude mice was evaluated by hematoxylin-eosin, immunohistochemical, and terminal deoxynucleotidyl transferase dUTP Nick-End Labeling. In BLCA, COL6A1 expression was downregulated. Moreover, the COL6A1 overexpression suppressed the viability, migration, and invasion, while promoting apoptosis of BLCA cell lines, with increased Caspase-3, Bax, and p53, and decreased Bcl-2. Conversely, silencing of COL6A1 promoted proliferation, migration, and invasion, while inhibiting apoptosis in BLCA cell lines. In vivo, COL6A1 inhibits tumor growth and progression. Fibrillin-1 (FBN1) was positively correlated with COL6A1 expression. COL6A1 could bind to FBN1 in BLCA cell lines. The expression of FBN1 in BLCA cell lines decreased after COL6A1 silencing, whereas COL6A1 overexpression upregulated FBN1 expression. COL6A1 was downregulated and exerted an inhibitory effect on the development of BLCA, and its expression was positively correlated with the expression of FBN1.
{"title":"COL6A1 Inhibits the Malignant Development of Bladder Cancer by Regulating FBN1.","authors":"Tineng Yang, Xiaoyang Peng, Xi Huang, Peng Cao, Hualei Chen","doi":"10.1007/s12013-024-01573-6","DOIUrl":"https://doi.org/10.1007/s12013-024-01573-6","url":null,"abstract":"<p><p>Bladder cancer (BLCA) is a prevalent malignancy worldwide with a high recurrence rate. Collagen Type VI Alpha 1 (COL6A1) plays a key role in several cancer types. In this study, we aimed to explore the role of COL6A1 in BLCA. COL6A1 expression in BLCA was determined using The Cancer Genome Atlas database and real-time quantitative polymerase chain reaction (RT-qPCR). Counting Kit-8, wound-healing, and transwell assays were used to assess the effect of COL6A1 on T24 and 5637 cells. Apoptosis in BLCA cell lines was explored using western blotting and flow cytometry. Co-immunoprecipitation was performed to determine interactions between proteins. The role of COL6A1 in tumor growth in nude mice was evaluated by hematoxylin-eosin, immunohistochemical, and terminal deoxynucleotidyl transferase dUTP Nick-End Labeling. In BLCA, COL6A1 expression was downregulated. Moreover, the COL6A1 overexpression suppressed the viability, migration, and invasion, while promoting apoptosis of BLCA cell lines, with increased Caspase-3, Bax, and p53, and decreased Bcl-2. Conversely, silencing of COL6A1 promoted proliferation, migration, and invasion, while inhibiting apoptosis in BLCA cell lines. In vivo, COL6A1 inhibits tumor growth and progression. Fibrillin-1 (FBN1) was positively correlated with COL6A1 expression. COL6A1 could bind to FBN1 in BLCA cell lines. The expression of FBN1 in BLCA cell lines decreased after COL6A1 silencing, whereas COL6A1 overexpression upregulated FBN1 expression. COL6A1 was downregulated and exerted an inhibitory effect on the development of BLCA, and its expression was positively correlated with the expression of FBN1.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370579","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}
Pub Date : 2024-10-04DOI: 10.1007/s12013-024-01583-4
Xiufang Du, Hongjie Yang, Xiaobei Kang, Changna Fu, Tao Yang
Endometriosis is a benign gynecological disorder characterized by the abnormal presence of endometrium-like cells, referred to as ectopic tissue, located outside the uterine cavity. Beyond the abnormal proliferation of endometrium-like tissues within and beyond the pelvic cavity, compelling scientific evidence underscores the crucial involvement of the NOD-like receptor NLRP3 inflammasome and pyroptosis in the pathogenesis of EMS. Our investigation has revealed a striking upregulation of the endogenous protein GATA-binding protein 6 (GATA6) in abdominal wall EMS. Notably, the knockdown of GATA6 significantly impaired the viability and migratory potential of primary ectopic endometrial stromal cells (EESCs) while also inhibiting crucial markers of pyroptosis, such as NLRP3, the gasdermin D N-terminal fragment (GSDMD-N), and reactive oxygen species (ROS) levels within these cells. Delving deeper into the underlying mechanisms, we discovered that suppressing GATA6 mitigated the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in EESCs. The administration of 740 Y-P, an agonist of the PI3K/AKT pathway, mitigated the inhibitive actions of GATA6 knockdown on EESCs' growth, migration, and pyroptosis, highlighting the intricate crosstalk between GATA6 and this intricate signaling cascade. In vivo experiments corroborated these findings, demonstrating that reduced GATA6 expression effectively restrained the growth of endometrial lesions and concurrently suppressed pyroptosis, accompanied by a dampening of PI3K/AKT signaling within these lesions. In summary, our study underscores the pivotal role of GATA6 in modulating the growth and pyroptosis of abdominal wall EMS through its regulation of the PI3K/AKT signaling pathway. Silencing GATA6 emerges as a promising approach to alleviate pyroptosis and potentially offers a novel therapeutic angle for managing abdominal wall EMS.
{"title":"Blocking GATA6 Alleviates Pyroptosis and Inhibits Abdominal Wall Endometriosis Lesion Growth Through Inactivating the PI3K/AKT Pathway.","authors":"Xiufang Du, Hongjie Yang, Xiaobei Kang, Changna Fu, Tao Yang","doi":"10.1007/s12013-024-01583-4","DOIUrl":"https://doi.org/10.1007/s12013-024-01583-4","url":null,"abstract":"<p><p>Endometriosis is a benign gynecological disorder characterized by the abnormal presence of endometrium-like cells, referred to as ectopic tissue, located outside the uterine cavity. Beyond the abnormal proliferation of endometrium-like tissues within and beyond the pelvic cavity, compelling scientific evidence underscores the crucial involvement of the NOD-like receptor NLRP3 inflammasome and pyroptosis in the pathogenesis of EMS. Our investigation has revealed a striking upregulation of the endogenous protein GATA-binding protein 6 (GATA6) in abdominal wall EMS. Notably, the knockdown of GATA6 significantly impaired the viability and migratory potential of primary ectopic endometrial stromal cells (EESCs) while also inhibiting crucial markers of pyroptosis, such as NLRP3, the gasdermin D N-terminal fragment (GSDMD-N), and reactive oxygen species (ROS) levels within these cells. Delving deeper into the underlying mechanisms, we discovered that suppressing GATA6 mitigated the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in EESCs. The administration of 740 Y-P, an agonist of the PI3K/AKT pathway, mitigated the inhibitive actions of GATA6 knockdown on EESCs' growth, migration, and pyroptosis, highlighting the intricate crosstalk between GATA6 and this intricate signaling cascade. In vivo experiments corroborated these findings, demonstrating that reduced GATA6 expression effectively restrained the growth of endometrial lesions and concurrently suppressed pyroptosis, accompanied by a dampening of PI3K/AKT signaling within these lesions. In summary, our study underscores the pivotal role of GATA6 in modulating the growth and pyroptosis of abdominal wall EMS through its regulation of the PI3K/AKT signaling pathway. Silencing GATA6 emerges as a promising approach to alleviate pyroptosis and potentially offers a novel therapeutic angle for managing abdominal wall EMS.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370578","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}
Infection occurs when pathogens penetrate tissues, reproduce, and trigger a host response to both the infectious agents and their toxins. A diverse array of pathogens, including viruses and bacteria, can cause infections. The host's immune system employs several mechanisms to combat these infections, typically involving an innate inflammatory response. Inflammation is a complex biological reaction that can affect various parts of the body and is a key component of the response to harmful stimuli. Sepsis arises when the body's response to infection leads to widespread damage to tissues and organs, potentially resulting in severe outcomes or death. The initial phase of sepsis involves immune system suppression. Early identification and targeted management are crucial for improving sepsis outcomes. Common treatment approaches include antibiotics, intravenous fluids, blood cultures, and monitoring urine output. This study explores the potential of exosome therapy in enhancing the management and alleviation of sepsis symptoms.
{"title":"The Therapeutic Potential of Exosome Therapy in Sepsis Management: Addressing Complications and Improving Outcomes\".","authors":"Payam Ali-Khiavi, Mahya Mohammadi, Sajjad Masoumi, Hossein Saffarfar, Reza Kheradmand, Ahmad Mobed, Faezeh Hatefnia","doi":"10.1007/s12013-024-01564-7","DOIUrl":"https://doi.org/10.1007/s12013-024-01564-7","url":null,"abstract":"<p><p>Infection occurs when pathogens penetrate tissues, reproduce, and trigger a host response to both the infectious agents and their toxins. A diverse array of pathogens, including viruses and bacteria, can cause infections. The host's immune system employs several mechanisms to combat these infections, typically involving an innate inflammatory response. Inflammation is a complex biological reaction that can affect various parts of the body and is a key component of the response to harmful stimuli. Sepsis arises when the body's response to infection leads to widespread damage to tissues and organs, potentially resulting in severe outcomes or death. The initial phase of sepsis involves immune system suppression. Early identification and targeted management are crucial for improving sepsis outcomes. Common treatment approaches include antibiotics, intravenous fluids, blood cultures, and monitoring urine output. This study explores the potential of exosome therapy in enhancing the management and alleviation of sepsis symptoms.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370581","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}
Pub Date : 2024-10-03DOI: 10.1007/s12013-024-01558-5
Huan Cheng, Zeyu Chen, Yong Wang, Chengjian Ji, Junqi Wang, Ninghong Song
Prostate cancer (PC) is the most frequently occurring cancer in men, characterized by the abnormal proliferation of cells within the prostate gland. This study explores the role of RNA binding motif protein 15B (RBM15B) in PC. RBM15B expression levels in PC patients were predicted using the Starbase database. The expression of RBM15B and proliferating cell nuclear antigen (PCNA) expression in PC cells was detected. Following RBM15B knockdown, cell proliferation assays were conducted. N6-methyladenosine (m6A) levels in PC cells were quantified, and RNA immunoprecipitation was performed to analyze the binding of m6A and YTH N-methyladenosine RNA binding protein 1 (YTHDF1) on PCNA mRNA. The stability of PCNA mRNA was assessed after treatment with actinomycin D. An in vivo nude mouse xenograft model was created to validate the role of RBM15B. The findings revealed the upregulation of RBM15B in PC. RBM15B knockdown resulted in decreased proliferation, colony formation, and EdU-positive cells. Mechanical analysis showed that RBM15B facilitated m6A modification of PCNA mRNA, leading to increasing m6A methylation. YTHDF1 bound to these m6A sites on PCNA mRNA, thus stabilizing it. Furthermore, PCNA overexpression mitigated the effects of RBM15B knockdown on PC cell proliferation. In conclusion, RBM15B promotes PC cell proliferation by enhancing the stability of PCNA mRNA through YTHDF1-mediated m6A modification.
{"title":"RBM15B Promotes Prostate Cancer Cell Proliferation via PCNA m6A Modification.","authors":"Huan Cheng, Zeyu Chen, Yong Wang, Chengjian Ji, Junqi Wang, Ninghong Song","doi":"10.1007/s12013-024-01558-5","DOIUrl":"https://doi.org/10.1007/s12013-024-01558-5","url":null,"abstract":"<p><p>Prostate cancer (PC) is the most frequently occurring cancer in men, characterized by the abnormal proliferation of cells within the prostate gland. This study explores the role of RNA binding motif protein 15B (RBM15B) in PC. RBM15B expression levels in PC patients were predicted using the Starbase database. The expression of RBM15B and proliferating cell nuclear antigen (PCNA) expression in PC cells was detected. Following RBM15B knockdown, cell proliferation assays were conducted. N6-methyladenosine (m6A) levels in PC cells were quantified, and RNA immunoprecipitation was performed to analyze the binding of m6A and YTH N-methyladenosine RNA binding protein 1 (YTHDF1) on PCNA mRNA. The stability of PCNA mRNA was assessed after treatment with actinomycin D. An in vivo nude mouse xenograft model was created to validate the role of RBM15B. The findings revealed the upregulation of RBM15B in PC. RBM15B knockdown resulted in decreased proliferation, colony formation, and EdU-positive cells. Mechanical analysis showed that RBM15B facilitated m6A modification of PCNA mRNA, leading to increasing m6A methylation. YTHDF1 bound to these m6A sites on PCNA mRNA, thus stabilizing it. Furthermore, PCNA overexpression mitigated the effects of RBM15B knockdown on PC cell proliferation. In conclusion, RBM15B promotes PC cell proliferation by enhancing the stability of PCNA mRNA through YTHDF1-mediated m6A modification.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363902","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}
Pub Date : 2024-10-01DOI: 10.1007/s12013-024-01563-8
Yan Ma, Hongjun Lin, Yunman Li, Zhuoling An
Rheumatoid arthritis (RA) is a systemic autoimmune disease that is prevalent worldwide and seriously threatens human health. RA-fibroblast-like synoviocytes (FLS) play important roles in almost all aspects of RA progression. This study aimed to study the effect of Amentoflavone (AMF), a polyphenol compound derived from extracts of Selaginella tamariscina, on the abnormal biological behaviors of RA-FLS. The immortalized human RA-FLS cell line (MH7A) was treated with AMF or transfected with small interfering RNAs (siRNAs) targeting peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1). Then, cell viability was detected by CCK-8 assay. EDU staining, wound healing and transwell assays were employed to measure the capacities of MH7A cell proliferation, migration and invasion. The levels of inflammatory factors were assessed using ELISA kits. Additionally, ferroptosis was analyzed by detecting Fe2+ content, lipid reactive oxygen species (ROS) level and expression of ferroptosis-related proteins. Pull-down assay was employed to verify the targeted binding of AMF to PIN1. Further, PIN1 overexpression or ferroptosis inhibitor Ferrostatin-1 (Fer-1) addition was conducted to elucidate the regulatory mechanism of AMF on PIN1 and ferroptosis. Results revealed that AMF intervention or PIN1 knockdown inhibited the proliferation, migration, invasion and inflammation in MH7A cells. AMF facilitated lipid peroxidation and ferroptosis in MH7A cells. Moreover, AMF targeted inhibition of PIN1 expression, and PIN1 overexpression restored the promoting effect of AMF on lipid peroxidation and ferroptosis in MH7A cells. Besides, Fer-1 reversed the impacts of AMF on the abnormal biological behaviors of MH7A cells. In summary, AMF induced ferroptosis to inhibit the proliferation, migration, invasion and inflammation in RA-FLS by inhibiting PIN1, providing a promising candidate for RA treatment.
{"title":"Amentoflavone Induces Ferroptosis to Alleviate Proliferation, Migration, Invasion and Inflammation in Rheumatoid Arthritis Fibroblast-like Synoviocytes by Inhibiting PIN1.","authors":"Yan Ma, Hongjun Lin, Yunman Li, Zhuoling An","doi":"10.1007/s12013-024-01563-8","DOIUrl":"https://doi.org/10.1007/s12013-024-01563-8","url":null,"abstract":"<p><p>Rheumatoid arthritis (RA) is a systemic autoimmune disease that is prevalent worldwide and seriously threatens human health. RA-fibroblast-like synoviocytes (FLS) play important roles in almost all aspects of RA progression. This study aimed to study the effect of Amentoflavone (AMF), a polyphenol compound derived from extracts of Selaginella tamariscina, on the abnormal biological behaviors of RA-FLS. The immortalized human RA-FLS cell line (MH7A) was treated with AMF or transfected with small interfering RNAs (siRNAs) targeting peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1). Then, cell viability was detected by CCK-8 assay. EDU staining, wound healing and transwell assays were employed to measure the capacities of MH7A cell proliferation, migration and invasion. The levels of inflammatory factors were assessed using ELISA kits. Additionally, ferroptosis was analyzed by detecting Fe<sup>2+</sup> content, lipid reactive oxygen species (ROS) level and expression of ferroptosis-related proteins. Pull-down assay was employed to verify the targeted binding of AMF to PIN1. Further, PIN1 overexpression or ferroptosis inhibitor Ferrostatin-1 (Fer-1) addition was conducted to elucidate the regulatory mechanism of AMF on PIN1 and ferroptosis. Results revealed that AMF intervention or PIN1 knockdown inhibited the proliferation, migration, invasion and inflammation in MH7A cells. AMF facilitated lipid peroxidation and ferroptosis in MH7A cells. Moreover, AMF targeted inhibition of PIN1 expression, and PIN1 overexpression restored the promoting effect of AMF on lipid peroxidation and ferroptosis in MH7A cells. Besides, Fer-1 reversed the impacts of AMF on the abnormal biological behaviors of MH7A cells. In summary, AMF induced ferroptosis to inhibit the proliferation, migration, invasion and inflammation in RA-FLS by inhibiting PIN1, providing a promising candidate for RA treatment.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142360885","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}
Pub Date : 2024-09-30DOI: 10.1007/s12013-024-01567-4
Li Qiu, Zhaoming Huang
Pediatric pneumonia is an inflammatory disease with a very high incidence. IGF binding protein 7 (IGFBP7) plays an important role in inflammatory diseases. However, the role of IGFBP7 in pediatric pneumonia and its mechanism have not been reported. Human embryonic lung (WI-38) cells were induced by lipopolysaccharide (LPS) to construct the cell inflammatory injury model. Subsequently, the expression of IGFBP7 was detected by qPCR and western blot. Next, IGFBP7 interference plasmid was constructed, and cell viability and apoptosis were detected by CCK8, flow cytometry and western blot. ELISA and other techniques were used to detect the inflammatory level. Autophagy and mitochondrial activities were detected by immunofluorescence and other techniques, and mitophagy-related proteins were detected by western blot. To further investigate the regulatory mechanism of IGFBP7, we administered cyclosporin A, a mitophagy inhibitor, and then detected apoptosis and inflammation. The expression of IGFBP7 was significantly increased in LPS-induced WI-38 cells. Interference with IGFBP7 expression in LPS-induced cells significantly increased cell activity, decreased apoptosis and cellular inflammation levels. During this process, mitophagy was enhanced. Further addition of cyclosporin A significantly reversed the protective effect of IGFBP7 knockdown. To be concluded, inhibition of IGFBP7 alleviates LPS-induced inflammation and apoptosis in WI-38 cells via enhancing mitophagy.
{"title":"Downregulation of IGFBP7 Alleviates LPS-induced Inflammation and Apoptosis in WI-38 Cells via Enhancing Mitophagy.","authors":"Li Qiu, Zhaoming Huang","doi":"10.1007/s12013-024-01567-4","DOIUrl":"https://doi.org/10.1007/s12013-024-01567-4","url":null,"abstract":"<p><p>Pediatric pneumonia is an inflammatory disease with a very high incidence. IGF binding protein 7 (IGFBP7) plays an important role in inflammatory diseases. However, the role of IGFBP7 in pediatric pneumonia and its mechanism have not been reported. Human embryonic lung (WI-38) cells were induced by lipopolysaccharide (LPS) to construct the cell inflammatory injury model. Subsequently, the expression of IGFBP7 was detected by qPCR and western blot. Next, IGFBP7 interference plasmid was constructed, and cell viability and apoptosis were detected by CCK8, flow cytometry and western blot. ELISA and other techniques were used to detect the inflammatory level. Autophagy and mitochondrial activities were detected by immunofluorescence and other techniques, and mitophagy-related proteins were detected by western blot. To further investigate the regulatory mechanism of IGFBP7, we administered cyclosporin A, a mitophagy inhibitor, and then detected apoptosis and inflammation. The expression of IGFBP7 was significantly increased in LPS-induced WI-38 cells. Interference with IGFBP7 expression in LPS-induced cells significantly increased cell activity, decreased apoptosis and cellular inflammation levels. During this process, mitophagy was enhanced. Further addition of cyclosporin A significantly reversed the protective effect of IGFBP7 knockdown. To be concluded, inhibition of IGFBP7 alleviates LPS-induced inflammation and apoptosis in WI-38 cells via enhancing mitophagy.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142338786","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}
Pub Date : 2024-09-29DOI: 10.1007/s12013-024-01552-x
Alma Karen Lomeli-Lepe, José Luis Castañeda-Cabral, Mónica E Ureña-Guerrero, Graciela Gudiño Cabrera, Silvia Josefina López-Pérez
Neuroinflammation and oxidative stress are important features in the pathogenesis and development of synucleinopathies, the glial activation and upregulation of pro-inflammatory and oxidative mediators induce alpha-synuclein (α-syn) accumulation. Recent studies have shown that bee venom (BV) has beneficial effects on symptoms of these neurodegenerative diseases. BV is known to exert anti-inflammatory and anti-oxidative effects. Here, we investigated the effects of BV over the different inflammatory and oxidative markers, and in the expression of α-syn and tyrosine hydroxylase (TH) in a lipopolysaccharide (LPS)-induced rat model of synucleinopathies. We examined whether BV (1.5 mg/kg by acupoint injection ST36 six times every 48 h) could change the α-syn and TH expression measured by western blotting, also, observed the activation of microglia and astrocytes by immunofluorescence, quantified the proinflammatory cytokines levels of tumoral necrosis factor-α (TNF-α) and Interleukin-1β (IL-1β) by enzyme-linked immunosorbent assay (ELISA), and estimated the lipid peroxidation and the activity of superoxide dismutase (SOD) and catalase (CAT) by colorimetric kits in LPS-treated rats (2.5 µg by a single dose intranigral injection) in substantia nigra (SN) and striatum (STR) brain areas. In the LPS-injected rat brain, BV treatment reduced α-syn levels and increased the TH levels. In addition, we observed lower microglia and astrocyte activation in SN and STR. Furthermore, BV decreases IL-1β and lipid peroxidation and increases the CAT activity in the STR. These results indicate that BV can restore the α-syn and TH levels possibly by the inhibition of LPS-induced neuroinflammation and oxidation, also, these results suggest that BV could be a promising treatment option for synucleinopathies.
{"title":"Bee Venom Reduces Early Inflammation and Oxidative Stress Associated with Lipopolysaccharide-induced Alpha-synuclein in the Substantia Nigra-striatum Axis.","authors":"Alma Karen Lomeli-Lepe, José Luis Castañeda-Cabral, Mónica E Ureña-Guerrero, Graciela Gudiño Cabrera, Silvia Josefina López-Pérez","doi":"10.1007/s12013-024-01552-x","DOIUrl":"https://doi.org/10.1007/s12013-024-01552-x","url":null,"abstract":"<p><p>Neuroinflammation and oxidative stress are important features in the pathogenesis and development of synucleinopathies, the glial activation and upregulation of pro-inflammatory and oxidative mediators induce alpha-synuclein (α-syn) accumulation. Recent studies have shown that bee venom (BV) has beneficial effects on symptoms of these neurodegenerative diseases. BV is known to exert anti-inflammatory and anti-oxidative effects. Here, we investigated the effects of BV over the different inflammatory and oxidative markers, and in the expression of α-syn and tyrosine hydroxylase (TH) in a lipopolysaccharide (LPS)-induced rat model of synucleinopathies. We examined whether BV (1.5 mg/kg by acupoint injection ST36 six times every 48 h) could change the α-syn and TH expression measured by western blotting, also, observed the activation of microglia and astrocytes by immunofluorescence, quantified the proinflammatory cytokines levels of tumoral necrosis factor-α (TNF-α) and Interleukin-1β (IL-1β) by enzyme-linked immunosorbent assay (ELISA), and estimated the lipid peroxidation and the activity of superoxide dismutase (SOD) and catalase (CAT) by colorimetric kits in LPS-treated rats (2.5 µg by a single dose intranigral injection) in substantia nigra (SN) and striatum (STR) brain areas. In the LPS-injected rat brain, BV treatment reduced α-syn levels and increased the TH levels. In addition, we observed lower microglia and astrocyte activation in SN and STR. Furthermore, BV decreases IL-1β and lipid peroxidation and increases the CAT activity in the STR. These results indicate that BV can restore the α-syn and TH levels possibly by the inhibition of LPS-induced neuroinflammation and oxidation, also, these results suggest that BV could be a promising treatment option for synucleinopathies.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142338784","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}
Pub Date : 2024-09-29DOI: 10.1007/s12013-024-01560-x
Fangfang Zhong, Yan Zeng, Jing Liu, Qulian Guo, Chunyan Liu, Wenjun Liu
The purpose of this study was to explore the antitumor effect and mechanism of Salvia miltiorrhiza injection (SMI) on acute myeloid leukemia (AML) cells in vitro and in vivo. Bioinformatics was used to detect c-Myc mRNA expression in AML patients in the Oncomine database. qRT‒PCR and western blotting were used to detect the mRNA and protein expression of c-Myc and HOXA5 in clinical samples. Different concentrations (6.25, 12.5, 25, 50 and 100 μg/mL) of SMI were added to KG1a and HL60 cells for 24, 48 and 72 h to determine the IC50 value of SMI. A CCK-8 assay was used to detect the effects of different concentrations of SMI and different treatment times on the proliferation of KG1a and HL60 cells. The indicated concentrations of SMI and SB203580 were used to treat KG1a and HL60 cells. The cell cycle distribution was determined by flow cytometry. The percentage of apoptotic cells was detected by Hoechst 33258 staining and flow cytometry. qRT‒PCR was performed to detect the mRNA expression of p38, c-Myc and HOXA5 in KG1a and HL60 cells. Western blotting was used to detect the protein expression of p38, p-p38, c-Myc, HOXA5, cCaspase 3 and cPARP in KG1a and HL60 cells. AutoDock software was used to analyze the molecular docking of the three main active components of SMI with c-Myc. AutoDock analysis revealed that the binding effect of molecular leisure was evaluated by binding energy, and a binding energy <-5 kcal/mol was considered good. SMI decreased the mRNA and protein expression of c-Myc and HOXA5. SMI significantly inhibited the proliferative activity of KG1a and HL60 cells and induced their apoptosis. However, SMI had no significant effect on the cell cycle distribution of KG1a and HL60 cells. With increasing SMI concentrations, the p-p38/p38 ratio increased, while the protein expression of c-Myc and HOXA5 decreased, and the protein expression of cCaspase and cPARP increased. However, SB203580 intervention in addition to SMI reversed these changes. Tanshinone IIA, cryptanshinone and salvianolic acid B can bind to multiple sites of c-Myc. In summary, SMI could be used for the treatment of acute leukemia, and its mechanism may be related to activation of the p38MAPK signaling pathway.
{"title":"Salvia Miltiorrhiza Injection Inhibited the Proliferation of AML Cells by Inducing Apoptosis through the p38MAPK Pathway.","authors":"Fangfang Zhong, Yan Zeng, Jing Liu, Qulian Guo, Chunyan Liu, Wenjun Liu","doi":"10.1007/s12013-024-01560-x","DOIUrl":"https://doi.org/10.1007/s12013-024-01560-x","url":null,"abstract":"<p><p>The purpose of this study was to explore the antitumor effect and mechanism of Salvia miltiorrhiza injection (SMI) on acute myeloid leukemia (AML) cells in vitro and in vivo. Bioinformatics was used to detect c-Myc mRNA expression in AML patients in the Oncomine database. qRT‒PCR and western blotting were used to detect the mRNA and protein expression of c-Myc and HOXA5 in clinical samples. Different concentrations (6.25, 12.5, 25, 50 and 100 μg/mL) of SMI were added to KG1a and HL60 cells for 24, 48 and 72 h to determine the IC<sub>50</sub> value of SMI. A CCK-8 assay was used to detect the effects of different concentrations of SMI and different treatment times on the proliferation of KG1a and HL60 cells. The indicated concentrations of SMI and SB203580 were used to treat KG1a and HL60 cells. The cell cycle distribution was determined by flow cytometry. The percentage of apoptotic cells was detected by Hoechst 33258 staining and flow cytometry. qRT‒PCR was performed to detect the mRNA expression of p38, c-Myc and HOXA5 in KG1a and HL60 cells. Western blotting was used to detect the protein expression of p38, p-p38, c-Myc, HOXA5, cCaspase 3 and cPARP in KG1a and HL60 cells. AutoDock software was used to analyze the molecular docking of the three main active components of SMI with c-Myc. AutoDock analysis revealed that the binding effect of molecular leisure was evaluated by binding energy, and a binding energy <-5 kcal/mol was considered good. SMI decreased the mRNA and protein expression of c-Myc and HOXA5. SMI significantly inhibited the proliferative activity of KG1a and HL60 cells and induced their apoptosis. However, SMI had no significant effect on the cell cycle distribution of KG1a and HL60 cells. With increasing SMI concentrations, the p-p38/p38 ratio increased, while the protein expression of c-Myc and HOXA5 decreased, and the protein expression of cCaspase and cPARP increased. However, SB203580 intervention in addition to SMI reversed these changes. Tanshinone IIA, cryptanshinone and salvianolic acid B can bind to multiple sites of c-Myc. In summary, SMI could be used for the treatment of acute leukemia, and its mechanism may be related to activation of the p38MAPK signaling pathway.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2024-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142338791","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}